+ All Categories
Home > Documents > 167060_Hemmes_thesis_compl... - UvA-DARE (Digital ...

167060_Hemmes_thesis_compl... - UvA-DARE (Digital ...

Date post: 07-Mar-2023
Category:
Upload: khangminh22
View: 0 times
Download: 0 times
Share this document with a friend
314
UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl) UvA-DARE (Digital Academic Repository) Lung-protective perioperative mechanical ventilation Hemmes, S.N.T. Publication date 2015 Document Version Final published version Link to publication Citation for published version (APA): Hemmes, S. N. T. (2015). Lung-protective perioperative mechanical ventilation. General rights It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons). Disclaimer/Complaints regulations If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible. Download date:01 Jun 2022
Transcript

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

Lung-protective perioperative mechanical ventilation

Hemmes, S.N.T.

Publication date2015Document VersionFinal published version

Link to publication

Citation for published version (APA):Hemmes, S. N. T. (2015). Lung-protective perioperative mechanical ventilation.

General rightsIt is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s)and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an opencontent license (like Creative Commons).

Disclaimer/Complaints regulationsIf you believe that digital publication of certain material infringes any of your rights or (privacy) interests, pleaselet the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the materialinaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letterto: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. Youwill be contacted as soon as possible.

Download date:01 Jun 2022

Lung-protective perioperative mechanical ventilation

Sabrine N

.T. Hem

mes

Lung-protective perioperative mechanical ventilation

Beyond the dangers of high tidals, on the perils of PEEP

Sabrine N.T. Hemmes

U I T N O D I G I N G

voor het bijwonen van de openbareverdediging van het proefschrift

LUNG-PROTECTIVE PERIOPERATIVEMECHANICAL VENTILATION

van

Sabrine Hemmes

Op vrijdag 11 december 2015om 13.00 uur

De Aula van de UvAOude Lutherse kerk

Singel 411, Amsterdam

Aansluitend bent u van harte uitgenodigd voor

de receptie ter plaatse

Paranimfen:Jet Heering

[email protected] - 14 77 53 37

Pieter Roel [email protected]

06 - 21 24 52 00

Sabrine HemmesVrolikstraat 232-1

1092TW [email protected]

Lung-protective perioperative mechanical ventilation

Beyond the dangers of high tidals, on the perils of PEEP

Sabrine N.T. Hemmes

COLOFONLung-protective perioperative mechanical ventilationAcademic thesis, University of Amsterdam, Amsterdam, The Netherlands

ISBN: 978-94-6233-149-5

Author: Sabrine N.T. HemmesLay-out: Barbara ten Brink Cover: Image by Heng Swee Lim - Website: http://www.ilovedoodle.comPrint: Gildeprint, Enschede, The Netherlands

Copyright © 2015 Sabrine N.T Hemmes, Amsterdam, The NetherlandsAll rights reserved. No part of this publication may be reproduced, stored, or transmitted in any form or by any means, without written permission of the author.

Financial support for the publication of this thesis was kindly provided by: Universiteit van Amsterdam, Abbvie, Edwards Lifesciences, Chipsoft

LUNG-PROTECTIVE PERIOPERATIVE MECHANICAL VENTILATION

ACADEMISCH PROEFSCHRIFT

ter verkrijging van de graad van doctor

aan de Universiteit van Amsterdam

op gezag van de Rector Magnificus

prof. dr. D.C. van den Boom

ten overstaan van een door het College voor Promoties ingestelde commissie,

in het openbaar te verdedigen in de Aula der Universiteit

op vrijdag 11 december 2015, te 13.00 uur

door Sabrine Nienke Tallechina Hemmes

geboren te Hilversum

PROMOTIECOMMISSIE

Promotores: prof. dr. M.J. Schultz Universiteit van Amsterdam prof. dr. dr. M.W. Hollmann Universiteit van Amsterdam

Overige leden: prof. dr. L.P.H.J. Aarts Universiteit van Leiden prof. dr. C. Boer Vrije Universiteit Amsterdam prof. dr. M.A. Boermeester Universiteit van Amsterdam prof. dr. G. Cinnella University of Foggia, Italy prof. dr. M.M. Levi Universiteit van Amsterdam prof. dr. W.S. Schlack Universiteit van Amsterdam prof. dr. M.B. Vroom Universiteit van Amsterdam

Faculteit der Geneeskunde

CONTENTS

Chapter 1 General introduction and outline of the thesis

Chapter 2 Intraoperative ventilatory strategies to prevent postoperative pulmonary complications – a metaanalysis

Current Opinion in Anesthesiology 2013

Chapter 3 Intraoperative protective mechanical ventilation for prevention of postoperative pulmonary complications – a comprehensive review of the role of tidal volume, positive end-expiratory pressure and lung recruitment manoeuvres

Anesthesiology 2015

Chapter 4 LAS VEGAS – Local assessment of ventilatory management during general anaesthesia for surgery and its effects on postoperative pulmonary complications: a prospective, observational, international, multicentre cohort study

European Journal of Anaesthesiology 2013

Chapter 5 Intraoperative ventilation strategies and patient outcomes following surgery: an international observational study (LAS VEGAS)

Manuscript submitted

Chapter 6 Protective ventilation with lower tidal volumes and high PEEP versus conventional ventilation with high tidal volume and low PEEP in patients under general anaesthesia for surgery: A systematic review and individual patient data metaanalysis

Anesthesiology 2015

Chapter 7 Rationale and study design of PROVHILO - a worldwide multicentre randomized controlled trial on protective ventilation during general anaesthesia for open abdominal surgery

Trials 2011

9

25

39

75

81

137

159

Chapter 8 High versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial

Lancet 2014

Chapter 9 Positive end-expiratory pressure during surgery - Authors' reply Lancet 2014

Chapter 10 Incidence of mortality and morbidity related to postoperative lung injury in patients who have undergone abdominal or thoracic surgery: a systematic review and metaanalysis

Lancet Respiratory Medicine 2014

Chapter 11 Positive end – expiratory pressure following coronary artery bypass grafting

Minerva Anestesiologica 2012

Chapter 12 Summary and general discussion

Appendices Nederlandse samenvatting

Contributing authors and affiliations

Publications

PhD Portfolio

Curriculum Vitae

Acknowledgements

177

215

223

251

267

284

296

302

304

308

310

Chapter 1

General introduction and outline of the thesis

Hemmes SNT, Schultz MJ

10

Prevention of postoperative pulmonary complications

Mechanical ventilation is frequently considered a simple but foremost harmless intervention in patients under general anaesthesia for surgery. Recent investigations, however, suggest that intraoperative ventilation has a strong potential to cause so called ventilator-associated lung injury.1 Of all patients undergoing ventilation during general anaesthesia for surgery, 5% will develop one or more postoperative pulmonary complications, that are associated with high morbidity and mortality.2, 3 There are several mechanisms through which intraoperative ventilation could cause ventilator–associated lung injury, as such contributing to development of postoperative pulmonary complications (fig. 1). First, positive pressure ventilation can overstretch patent alveoli causing damage in those parts of the lung that are aerated during the whole breath cycle (fig. 1A & 1C).4, 5 Second, repeated opening and closing of alveoli that collapse at the end of expiration is associated with increased shear stress, known to cause epithelial destruction (fig. 1B & 1D).6-8 Third, hyperoxia can result in absorption atelectasis, and cause injury of cellular structures through increased production of reactive oxygen species (ROS) (fig. 1E).9 All these harmful effects are suggested to be preventable through the use of lung–protective ventilator settings, using low tidal volumes for prevention of overdistension,10,11 use of positive end–expiratory pressure (PEEP)12, 13 with or without so–called recruitment manoeuvres to prevent repeated opening and closing,14, 15 and low oxygen fractions (FiO2) preventing atelectasis and ROS production.16 These insights have led to a paradigm shift from supranormal intraoperative ventilation, with large tidal volumes to prevent atelectasis and high levels of FiO2 to maximize oxygenation, to safer ventilation, using lower levels of tidal volumes, higher levels of PEEP and lower arterial oxygen thresholds.

Tidal volumes

Low tidal volumes in animal studiesThe harmful effects of high tidal volumes were first recognized in animal studies of ventilation.17 In these preclinical studies lungs of animals were either challenged with injurious ventilation strategies using different tidal volumes alone, or in combination with other challenges such as intratracheal instillation of lipopolysaccharide or live bacteria.18 More or less they all showed that the extent of alveolar damage and pulmonary oedema depends on the size of tidal volumes used.17, 18

Low tidal volumes in patients with ARDSTraditionally patients were ventilated with large tidal volumes of 10 to 15 mL/kg predicted body weight (PBW). These volumes far exceeded the range of normal tidal volumes in healthy subjects in rest (7 to 8 mL/kg PBW). The rationale was to prevent atelectasis, as such optimizing oxygenation and ventilation.19 Randomized controlled clinical trials in critically ill patients with the acute respiratory distress syndrome (ARDS), however, showed large tidal volumes to be harmful.20-24 Two metaanalyses convincingly confirmed that ventilation with low tidal volumes in patients with ARDS is associated with improved survival.25, 26 Consequently, nowadays ventilation with low tidal volumes is standard of care in these patients.27, 28

General introduction

Chap

ter

1

11

Low tidal volumes in critically ill patients without ARDSThe finding that ventilation with low tidal volumes benefits patients with ARDS evoked interest in lung–protective ventilation in critically ill patients who need ventilation for other reasons than ARDS, for example comatose patients with neurologic damage and patients after major cardiac surgery. One randomized controlled trial comparing ventilation with low tidal volumes (6 mL/kg PBW) to ventilation with high tidal volumes (10 mL/kg PBW) indeed suggested benefit from low tidal volumes, as it seemed to reduce the incidence of ARDS.29 These findings were confirmed in a series of metaanalyses.18, 30, 31 In addition, these analyses revealed that ventilation with low tidal volumes was associated with earlier liberation from the ventilator. Even though a substantial reduction in tidal volume size is seen in recent years,27, 28 lung–protective ventilation using low tidal volumes is not yet considered standard of care for critically ill patients who need ventilatory support for reasons other than ARDS.

Low tidal volumes during intraoperative ventilationSeveral small clinical trials of intraoperative ventilation suggested that tidal volume reduction could reduce local production of inflammatory mediators and possibly improve pulmonary mechanics.32-34 A large retrospective trial showed that pressure- and volume–limited ventilation during general surgery decreases the development of postoperative respiratory complications.35 Recently, three randomized controlled trials provided more robust evidence for benefit from this ventilation strategy.36-38 An Italian single–centre trial showed that a ventilation strategy using tidal volumes of 7 mL/kg PBW compared to ventilation with tidal volumes of 9 mL/kg PBW during abdominal surgery was associated with superior postoperative pulmonary function.36 A French multicentre trial found that in patients undergoing abdominal surgery a ventilation strategy with reduced tidal volumes of 6 mL/kg PBW compared to tidal volumes of 12 mL/kg PBW was associated with a decreased incidence of postoperative complications by almost 65%.37 One Chinese trial in patients undergoing spinal fusion reported an even more impressive benefit of tidal volume reduction from 12 to 6 mL/kg PBW on postoperative pulmonary complications.38 Contrasting to these results, one retrospective study showed that use of low tidal volume ventilation (6 to 8 mL/kg PBW) is associated with increased postoperative mortality, though the authors claim this to be caused by insufficient levels of PEEP.39 Despite the suggestion that low tidal volume ventilation in surgery patients is increasingly accepted,40, 41 recent studies show imperfect implementation of this strategy in the operation room.42-45

Positive end–expiratory pressure

Positive end–expiratory pressure in animal studiesSeveral studies in animals with lung injury have shown that ventilation with PEEP compared to ventilation without PEEP improves oxygenation and lung mechanics, and prevents formation of lung edema.5, 17, 46 Similar results came from studies in animals without lung injury. Ventilation with PEEP in combination with low tidal volumes attenuated local production of inflammatory mediators,47-52 lung edema,48, 51 and cell injury.47-52 One important shortcoming of PEEP, however, is that it could cause overdistension of the lung parts that remain aerated during the complete breath cycle.53 In addition, use of higher levels of PEEP could compromise the circulation.54

12

Figure 1. Mechanisms through which intraoperative ventilation could cause ventilator–associated lung injuryA) Ventilation at high lung volumes result in overdistention of the lung and hyperinflation may cause gross barotrauma (air leaks), but can also cause an increase in pulmonary oedema; B) ventilation at low lung volumes causes repeated opening and closing of alveoli that collapse at the end of expiration, resulting in increased shear stress and lung injury (atelectrauma). Collapse of large regions of the lung during ventilation at low lung volumes cause lung inhomogeneity; C) ventilation at too high levels of PEEP can aggravate overdistention of lung tissue at end-expiration; D) ventilation at low levels of PEEP increases formation of atelectasis and lung inhomogeneity; E) high levels of fractional inspired oxygen (FiO2) can increase the production of reactive oxygen species (ROS), which have a direct toxic effect on lung cells. Too high levels of FiO2 also increases the risk of resorption atelectasis; F) these mechanical and chemical stressors cause structural and biological changes in the alveoli. Inflammatory mediators are released in the lung and recruit neutrophils. They also cause changes that promote pulmonary fibrosis. The increase in alveolar-capillary permeability causes an increase in pulmonary oedema, but also facilitate translocation of mediators and bacteria to the systemic circulation; G) these structural and biological changes result in lung injury, which can cause an increase in postoperative pulmonary complications and worse clinical outcome with increased length of hospital stay and higher incidence of mortality (H)

Positive end–expiratory pressure in patients with ARDSThree randomized controlled trials in patients with ARDS failed to show an effect of higher levels of PEEP on survival.55-57 One metaanalysis using the individual patient data of these three trials, however, showed survival benefit in patients with more severe ARDS.58 Consequently, nowadays most clinicians use higher levels of PEEP (10 cm H2O and higher) in patients with moderate or severe ARDS.27

General introduction

Chap

ter

1

13

Positive end–expiratory pressure in critically ill patients without ARDSIn critical care patients without ARDS, there is limited evidence for benefit of PEEP.59, 60 One randomized controlled trial in patients at risk for ARDS showed no difference between a strategy using PEEP (5 to 8 cm H2O) and a strategy using a minimal levels of PEEP for adequate oxygenation with regard to later development of ARDS or mortality.59 This was confirmed in another trial in patients without ARDS, but in this trial use of higher levels of PEEP was associated with a lower incidence of ventilator–associated pneumonia.60 In the randomized controlled trial comparing low and high tidal volumes (6 ml/kg PBW versus 10 ml/kg PBW) in patients without ARDS mentioned above,29 an independent association between higher levels of PEEP and development of ARDS was found. In the postoperative phase there is also no clear evidence for benefit from PEEP. Indeed, while PEEP improves pulmonary compliance and arterial oxygenation, these effects only last in the first hours after surgery.61 This is also true for the prevention of atelectasis.62 In one trial in patients after cardiac surgery in which PEEP was titrated on the best achievable PaO2 level no sustained benefit was found.63 In general, intensive care unit clinicians now use PEEP levels between 4 and 7 cm H2O in critically ill patients who need ventilation for other reasons than ARDS,28 though the best level of PEEP for these patients remains unclear.

Positive end–expiratory pressure during intraoperative ventilationInduction of anaesthesia induces atelectasis,64 increasing ventilation–perfusion mismatch and suboptimal oxygenation.65 Intraoperative use of PEEP, with or without recruitment manoeuvres, is suggested to prevent atelectasis and repeated opening and closing of lung tissue.65 14 Indeed, use of higher levels of PEEP seems to improve oxygenation and respiratory mechanics in a wide range of patient populations and surgical settings.15, 66-74 However, in most trials of intraoperative PEEP, recruitment of lung tissue was not maintained in the postoperative period at the post anaesthesia care unit (PACU).72, 73

Benefit of higher levels of PEEP on postoperative outcome was suggested in the three randomized controlled trials of lung–protective intraoperative ventilation mentioned above, where use of low tidal volumes was actually combined with higher levels of PEEP with recruitment maneuvers.36-38 It is difficult, if not impossible to conclude what prevented postoperative complications: the use of low tidal volumes or the high levels of PEEP, or recruitment manoeuvres, or altogether. Despite this, both low tidal volumes and high levels of PEEP with recruitment manoeuvres are suggested to be beneficial.75 A recent large retrospective study confirms this suggestion, showing that both low tidal volumes (< 10 mL/kg) and higher PEEP levels (≥ 5 cmH2O) are independently associated with a decreased risk of postoperative respiratory complications.35

The lack of sufficient evidence for benefit of higher levels of PEEP during surgery is mirrored in the remarkable variation in use of intraoperative PEEP varying from 17% to as high as 82% of recently reported series.40-42, 44, 45

14

Oxygen fractions

Oxygen fractions in animal studiesThe potentially toxic effects of high fractions of inspired oxygen (FiO2) have long been known from animal studies. Mice exposed to hyperoxia develop a condition similar to ARDS, which is at least in part dependent on an increased production of reactive oxygen species (ROS) by mitochondria.76, 77 Hyperoxia could further cause atelectasis, tracheobronchitis, interstitial fibrosis, protein leakage and neutrophil infiltration.78-80 In spontaneous breathing rodents with pneumonia, hyperoxia has been shown to contribute to bacterial spread beyond the lungs,81 lung injury and even lethality.82 More cytokine production and increased lung injury was found in experiments in ventilated rodents with hyperoxia during injurious ventilation (tidal volume > 20 mL/kg).83-85

Oxygen fractions in patients with ARDSIn human lungs high FiO2 can also accelerate the production of ROS, which overwhelms natural anti–oxidant defences and injures cellular structures in the lung.9,16,86,87 Furthermore, hyperoxia can cause derecruitment of lung tissue by resorption atelectasis.88 Critically ill patients with lung injury are possibly more prone to the harmful pulmonary effects of oxygen toxicity, which can coincide with the primary pulmonary injury (e.g., ARDS, pneumonia) and ventilator–associated lung injury.77,89,90 One small trial found that 100% compared to 60% FiO2 increased development of atelectasis in patients with ARDS, which was prevented by application of higher levels of PEEP.91 However, clinical trials examining the effect of hyperoxia on the development of lung injury in patients with ARDS are lacking.

Oxygen fractions in critically ill patients without ARDSIn critical care patients who need ventilatory support for reasons other than ARDS, an association between hyperoxia and mortality was found in ventilated patients,89 patients after cardiac arrest,92 and patients with traumatic brain injury,93 or stroke.94 However, other studies did not reveal such associations.90, 95-97 For example, two recent metaanalyses investigating the effect of high FiO2 in critical care patients on survival showed mixed results.98, 99 One metaanalysis did not find a significant association in the general ICU,98 while another metaanalysis of pooled data from all critically ill patients suggested arterial hyperoxia to increase the risk of mortality.99 In subset analyses, hyperoxia was associated with decreased survival in patients after cardiac arrest, traumatic brain injury, and stroke.98 In patients after cardiac arrest a dose–dependent association between hyperoxia and patient outcome was found.99,100 Notably, arterial hyperoxia decreases coronary blood flow and cardiac output, increases systemic vascular resistance, and contributes to reperfusion injury in patients with myocardial infarction.101-104 A recent randomized controlled trial in patients with myocardial infarction indeed clearly showed that supplemental oxygen increased myocardial injury.105 Research on the effect of FiO2 on the development of lung injury in patients ventilated for other reasons than ARDS, however, is currently unavailable. Despite the lack of evidence, current guidelines in critically ill patients aim at PaO2 levels around 55–80 mm Hg.55,106

Oxygen fractions during intraoperative ventilationAnaesthesiologists use high FiO2 during pre–oxygenation and denitrogenation to prolong the apnoea tolerance time107 and during intraoperative ventilation to correct for arterial hypoxemia

General introduction

Chap

ter

1

15

induced by ventilation–perfusion mismatches caused by alveolar collapse.64 High FiO2 (> 80%) increases the incidence of resorption atelectasis, which not only augments atelectasis formation after induction,107,108 but also directly before emergence from anaesthesia in the post–oxygenation phase, annulling the open lung created during intraoperative ventilation.109 At the same time, there is a risk of hyperoxia–induced injury to the lungs. The available trials and metaanalyses on perioperative hyperoxia focused on the beneficial effect of high FiO2 on postoperative nausea and vomiting110-112 and postoperative wound infections.113-118 A large trial on postoperative wound infections investigated the effect of 80% compared to 30% oxygen during surgery on development of postoperative pulmonary complications as secondary endpoint and found no difference in incidence of atelectasis, pneumonia, and respiratory failure.117 A clinical trial in obese patients showed worse postoperative lung function in patients receiving FiO2 during ventilation.119 One metaanalysis found no difference in presence or absence of atelectasis or postoperative gas exchange during intraoperative ventilation with either high or low FiO2.120 A large recent metaanalysis, however, suggested that hyperoxia was not associated with increased 30–day mortality.121 Sufficiently powered clinical trials on lung injury and postoperative pulmonary complications due to high FiO2 are lacking.

Aims of this thesis

This thesis is a collection of investigations that focused on several aspects of perioperative ventilation, specifically ventilation practice and the associations between ventilator settings and the effects on postoperative pulmonary complications and outcome. The main interest was on PEEP. We hypothesized that the use of higher PEEP and recruitment manoeuvres would protect against development of postoperative pulmonary complications during intraoperative ventilation.

The specific aims of this thesis were:

1. To investigate the effect of intraoperative use of PEEP and recruitment manoeuvres on occurrence of postoperative pulmonary complications during low tidal volume ventilation during open abdominal surgery.

2. To determine the association between intraoperative use of high tidal volumes, PEEP and recruitment manoeuvres, and the occurrence of postoperative pulmonary complications.

3. To investigate the effects of development of postoperative lung injury on postoperative clinical course and mortality.

4. To examine the effects of different levels of PEEP during postoperative ventilation after coronary artery bypass grafting on the duration to extubation.

16

Outline of this thesis

The following chapters in this thesis report on observational studies, clinical trials and metaanalyses that reported on several aspects of lung–protective perioperative ventilation, including effects of tidal volume size and level of PEEP.

Chapter 2 provides the results of a metaanalysis of eight clinical trials examining the effects of intraoperative ventilator settings on postoperative outcome of non–cardiac surgery patients. We hypothesized that use of low tidal volumes and/or PEEP with or without recruitment manoeuvres could prevent postoperative pulmonary complications, and as such improving postoperative outcome. In this metaanalysis we tried to separate the effects of tidal volume and PEEP manipulations.

Chapter 3 constitutes a comprehensive review of the literature on predictive models of postoperative pulmonary complications, the pathophysiology of ventilation–induced lung injury, and protective ventilation strategies, including the respective roles of tidal volume size, the level of PEEP and the use of recruitment manoeuvres. In this review we propose an algorithm for protective intraoperative mechanical ventilation.

In Chapter 4 and chapter 5 we describe the design and the results, respectively, of the ‘Local Assessment of Ventilatory Management during General Anaesthesia for Surgery’–study (LAS VEGAS), a prospective observational cohort study designed to assess intraoperative ventilation practice in Europe and the America’s, and to test the hypothesis that certain ventilator settings, especially high tidal volumes and low PEEP levels, are associated with the occurrence of postoperative pulmonary complications.

In Chapter 6 we show the results of a metaanalysis using individual patient data from 15 randomized controlled trials of intraoperative ventilation. We hypothesized that intraoperative ventilation with lower tidal volumes protects against postoperative pulmonary complications, and that use of higher levels of PEEP adds to the beneficial effects of lower tidal volumes.

Chapter 7 and chapter 8 constitute the design and results of the PROVHILO trial (High versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery), a randomized controlled trial of intraoperative ventilation for open abdominal surgery. In chapter 9 entails letters with comments on PROVHILO written by peers, as well as our Author’s reply. In this trial patients were randomized to ventilation with high levels of PEEP (12 cm H2O) with recruitment manoeuvres or low levels of PEEP (0 to 2 cm H2O) without recruitment manoeuvres. We hypothesized that a ventilation strategy with high levels of PEEP and recruitment manoeuvres would protect against development of postoperative pulmonary complications.

In Chapter 10 we describe the results of another metaanalysis, using individual patient data from 12 clinical investigations of intraoperative ventilation. We hypothesized that the occurrence of postoperative lung injury was associated with a worse outcome, and that postoperative outcome would depend on intraoperative ventilation settings.

General introduction

Chap

ter

1

17

In Chapter 11 we present the results of a secondary analysis of two randomized controlled trials of postoperative ventilation in patients undergoing cardiac surgery, in which we determined the effects of PEEP manipulations on pulmonary compliance and gas exchange in the first hours of weaning from mechanical ventilation and time on the ventilator.122, 123 We hypothesized that higher levels of PEEP would improve pulmonary function, but not to be associated with a shorter duration of postoperative ventilation.

This thesis ends with a summary of the abovementioned studies and a general discussion in chapter 12, with Dutch translation in chapter 13.

18

References1. Tremblay LN, Slutsky AS. Ventilator-induced lung injury: from the bench to the bedside. Intensive care medicine 2006;

32(1): 24-332. Canet J, Gallart L, Gomar C, et al. Prediction of Postoperative Pulmonary Complications in a Population-based Surgical

Cohort. Anesthesiology 2010; 113(6): 1338-503. Mazo V, Sabate S, Canet J, et al. Prospective external validation of a predictive score for postoperative pulmonary

complications. Anesthesiology 2014; 121(2): 219-314. Chiumello D, Carlesso E, Cadringher P, et al. Lung stress and strain during mechanical ventilation for acute respiratory

distress syndrome. American journal of respiratory and critical care medicine 2008; 178(4): 346-555. Dreyfuss D, Saumon G. Ventilator-induced lung injury: lessons from experimental studies. American journal of respiratory

and critical care medicine 1998; 157(1): 294-3236. Mead J, Takishima T, Leith D. Stress distribution in lungs: a model of pulmonary elasticity. J Appl Physiol 1970; 28(5):

596-6087. Chu EK, Whitehead T, Slutsky AS. Effects of cyclic opening and closing at low- and high-volume ventilation on

bronchoalveolar lavage cytokines. Critical Care Medicine 2004; 32(1): 168-748. Albert RK. The role of ventilation-induced surfactant dysfunction and atelectasis in causing acute respiratory distress

syndrome. American journal of respiratory and critical care medicine 2012; 185(7): 702-89. Kallet RH, Matthay MA. Hyperoxic acute lung injury. Respiratory Care 2013; 58(1): 123-4110. Plataki M, Hubmayr RD. The physical basis of ventilator-induced lung injury. Expert Rev Respir Med 2010; 4(3): 373-8511. Rocco PR, Dos Santos C, Pelosi P. Pathophysiology of ventilator-associated lung injury. Current Opinion in Anaesthesiology

2012; 25(2): 123-3012. Neumann P, Rothen HU, Berglund JE, Valtysson J, Magnusson A, Hedenstierna G. Positive end-expiratory pressure

prevents atelectasis during general anaesthesia even in the presence of a high inspired oxygen concentration. Acta Anaesthesiologica Scandinavica 1999; 43(3): 295-301

13. Rusca M, Proietti S, Schnyder P, et al. Prevention of atelectasis formation during induction of general anesthesia. Anesthesia and Analgesia 2003; 97(6): 1835-9

14. Lachmann B. Open up the lung and keep the lung open. Intensive Care Medicine 1992; 18(6): 319-2115. Reinius H, Jonsson L, Gustafsson S, et al. Prevention of atelectasis in morbidly obese patients during general anesthesia

and paralysis: a computerized tomography study. Anesthesiology 2009; 111(5): 979-8716. Bhandari V, Elias JA. Cytokines in tolerance to hyperoxia-induced injury in the developing and adult lung. Free radical

biology & medicine 2006; 41(1): 4-1817. Dreyfuss D, Soler P, Basset G, Saumon G. High inflation pressure pulmonary edema. Respective effects of high airway

pressure, high tidal volume, and positive end-expiratory pressure. The American review of respiratory disease 1988; 137(5): 1159-64

18. Serpa Neto A, Nagtzaam L, Schultz MJ. Ventilation with lower tidal volumes for critically ill patients without the acute respiratory distress syndrome: a systematic translational review and metaanalysis. Current opinion in critical care 2014; 20(1): 25-32

19. Marini JJ. Evolving concepts in the ventilatory management of acute respiratory distress syndrome. Clinics in chest medicine 1996; 17(3): 555-75

20. Amato MB, Barbas CS, Medeiros DM, et al. Effect of a protective-ventilation strategy on mortality in the acute respiratory distress syndrome. NEJM 1998; 338(6): 347-54

21. Brochard L, Roudot-Thoraval F, Roupie E, et al. Tidal volume reduction for prevention of ventilator-induced lung injury in acute respiratory distress syndrome. The Multicenter Trail Group on Tidal Volume reduction in ARDS. American journal of respiratory and critical care medicine 1998; 158(6): 1831-8

22. Stewart TE, Meade MO, Cook DJ, et al. Evaluation of a ventilation strategy to prevent barotrauma in patients at high risk for acute respiratory distress syndrome. Pressure- and Volume-Limited Ventilation Strategy Group. NEJM 1998; 338(6): 355-61

23. Brower RG, Shanholtz CB, Fessler HE, et al. Prospective, randomized, controlled clinical trial comparing traditional versus reduced tidal volume ventilation in acute respiratory distress syndrome patients. Critical care medicine 1999; 27(8): 1492-8

24. ARDSnet. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. The Acute Respiratory Distress Syndrome Network. NEJM 2000; 342(18): 1301-8

25. Putensen C, Theuerkauf N, Zinserling J, Wrigge H, Pelosi P. Metaanalysis: ventilation strategies and outcomes of the acute respiratory distress syndrome and acute lung injury. Ann Intern Med 2009; 151(8): 566-76

26. Burns KE, Adhikari NK, Slutsky AS, et al. Pressure and volume limited ventilation for the ventilatory management of

General introduction

Chap

ter

1

19

patients with acute lung injury: a systematic review and metaanalysis. PLoS One 2011; 6(1): e1462327. Needham DM, Colantuoni E, Mendez-Tellez PA, et al. Lung protective mechanical ventilation and two year survival in

patients with acute lung injury: prospective cohort study. BMJ 2012; 344: e212428. Esteban A, Frutos-Vivar F, Muriel A, et al. Evolution of mortality over time in patients receiving mechanical ventilation.

American journal of respiratory and critical care medicine 2013; 188(2): 220-3029. Determann RM, Royakkers A, Wolthuis EK, et al. Ventilation with lower tidal volumes as compared with conventional tidal

volumes for patients without acute lung injury: a preventive randomized controlled trial. Critical Care 2010; 14(1): R130. Serpa Neto A, Simonis FD, Barbas CS, et al. Association between tidal volume size, duration of ventilation, and sedation

needs in patients without acute respiratory distress syndrome: an individual patient data metaanalysis. Intensive Care Medicine 2014; 40(7): 950-7

31. Serpa Neto A, Cardoso SO, Manetta JA, et al. Association between use of lung-protective ventilation with lower tidal volumes and clinical outcomes among patients without acute respiratory distress syndrome: a metaanalysis. JAMA 2012; 308(16): 1651-9

32. Zupancich E, Paparella D, Turani F, et al. Mechanical ventilation affects inflammatory mediators in patients undergoing cardiopulmonary bypass for cardiac surgery: a randomized clinical trial. The Journal of thoracic and cardiovascular surgery 2005; 130(2): 378-83

33. Chaney MA, Nikolov MP, Blakeman BP, Bakhos M. Protective ventilation attenuates postoperative pulmonary dysfunction in patients undergoing cardiopulmonary bypass. Journal of cardiothoracic and vascular anesthesia 2000; 14(5): 514-8

34. Michelet P, D’Journo XB, Roch A, et al. Protective ventilation influences systemic inflammation after esophagectomy: a randomized controlled study. Anesthesiology 2006; 105(5): 911-9

35. Ladha K, Vidal Melo MF, McLean DJ, et al. Intraoperative protective mechanical ventilation and risk of postoperative respiratory complications: hospital based registry study. BMJ 2015; 351: h3646

36. Severgnini P, Selmo G, Lanza C, et al. Protective mechanical ventilation during general anesthesia for open abdominal surgery improves postoperative pulmonary function. Anesthesiology 2013; 118(6): 1307-21

37. Futier E, Constantin JM, Paugam-Burtz C, et al. A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. NEJM 2013; 369(5): 428-37

38. Ge Y, Yuan L, Jiang X, Wang X, Xu R, Ma W. [Effect of lung protection mechanical ventilation on respiratory function in the elderly undergoing spinal fusion]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2013; 38(1): 81-5

39. Levin MA, McCormick PJ, Lin HM, Hosseinian L, Fischer GW. Low intraoperative tidal volume ventilation with minimal PEEP is associated with increased mortality. British journal of anaesthesia 2014; 113(1): 97-108

40. Treschan TA, Schaefer MS, Subasi L, Kaisers W, Schultz MJ, Beiderlinden M. Evolution of ventilator settings during general anaesthesia for neurosurgery: An observational study in a German centre over 15 years. European journal of anaesthesiology 2015; Jan 13 [Epub ahead of print]

41. Wanderer JP, Ehrenfeld JM, Epstein RH, et al. Temporal trends and current practice patterns for intraoperative ventilation at U.S. academic medical centers: a retrospective study. BMC Anesthesiology 2015; 15: 40

42. Jaber S, Coisel Y, Chanques G, et al. A multicentre observational study of intra-operative ventilatory management during general anaesthesia: tidal volumes and relation to body weight. Anaesthesia 2012; 67(9): 999-1008

43. Lellouche F, Dionne S, Simard S, Bussieres J, Dagenais F. High tidal volumes in mechanically ventilated patients increase organ dysfunction after cardiac surgery. Anesthesiology 2012; 116(5): 1072-82

44. Hess DR, Kondili D, Burns E, Bittner EA, Schmidt UH. A 5-year observational study of lung-protective ventilation in the operating room: a single-center experience. J Crit Care 2013; 28(4): 533 e9-15

45. Karalapillai D, Weinberg L, Galtieri J, et al. Current ventilation practice during general anaesthesia: a prospective audit in Melbourne, Australia. BMC Anesthesiology 2014; 14: 85

46. Webb HH, Tierney DF. Experimental pulmonary edema due to intermittent positive pressure ventilation with high inflation pressures. Protection by positive end-expiratory pressure. The American review of respiratory disease 1974; 110(5): 556-65

47. Vobruba V, Klimenko OV, Kobr J, et al. Effects of high tidal volume mechanical ventilation on production of cytokines, iNOS, and MIP-1beta proteins in pigs. Experimental lung research 2013; 39(1): 1-8

48. Hegeman MA, Hemmes SN, Kuipers MT, et al. The extent of ventilator-induced lung injury in mice partly depends on duration of mechanical ventilation. Critical care research and practice 2013; 2013: 435236

49. Theroux MC, Fisher AO, Horner LM, et al. Protective ventilation to reduce inflammatory injury from one lung ventilation in a piglet model. Paediatric anaesthesia 2010; 20(4): 356-64

50. Wilson MR, Choudhury S, Goddard ME, O’Dea KP, Nicholson AG, Takata M. High tidal volume upregulates intrapulmonary cytokines in an in vivo mouse model of ventilator-induced lung injury. Journal of applied physiology 2003; 95(4): 1385-93

51. Ota S, Nakamura K, Yazawa T, et al. High tidal volume ventilation induces lung injury after hepatic ischemia-reperfusion.

20

American journal of physiology Lung cellular and molecular physiology 2007; 292(3): L625-3152. Fanelli V, Mascia L, Puntorieri V, et al. Pulmonary atelectasis during low stretch ventilation: “open lung” versus “lung

rest” strategy. Critical care medicine 2009; 37(3): 1046-53. Bellardine Black CL, Hoffman AM, Tsai LW, et al. Relationship between dynamic respiratory mechanics and disease

heterogeneity in sheep lavage injury. Critical care medicine 2007; 35(3): 870-854. Luce JM. The cardiovascular effects of mechanical ventilation and positive end-expiratory pressure. Jama 1984; 252(6):

807-1155. Brower RG, Lanken PN, MacIntyre N, et al. Higher versus lower positive end-expiratory pressures in patients with the

acute respiratory distress syndrome. The New England journal of medicine 2004; 351(4): 327-3656. Meade MO, Cook DJ, Guyatt GH, et al. Ventilation strategy using low tidal volumes, recruitment maneuvers, and high

positive end-expiratory pressure for acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. JAMA 2008; 299(6): 637-45

57. Mercat A, Richard JC, Vielle B, et al. Positive end-expiratory pressure setting in adults with acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. JAMA 2008; 299(6): 646-55

58. Briel M, Meade M, Mercat A, et al. Higher vs lower positive end-expiratory pressure in patients with acute lung injury and acute respiratory distress syndrome: systematic review and metaanalysis. JAMA 2010; 303(9): 865-73

59. Pepe PE, Hudson LD, Carrico CJ. Early application of positive end-expiratory pressure in patients at risk for the adult respiratory-distress syndrome. NEJM 1984; 311(5): 281-6

60. Manzano F, Fernandez-Mondejar E, Colmenero M, et al. Positive-end expiratory pressure reduces incidence of ventilator-associated pneumonia in nonhypoxemic patients. Critical Care Medicine 2008; 36(8): 2225-31

61. Ranieri VM, Vitale N, Grasso S, et al. Time-course of impairment of respiratory mechanics after cardiac surgery and cardiopulmonary bypass. Critical Care Medicine 1999; 27(8): 1454-60

62. Marvel SL, Elliott CG, Tocino I, Greenway LW, Metcalf SM, Chapman RH. Positive end-expiratory pressure following coronary artery bypass grafting. Chest 1986; 90(4): 537-41

63. Celebi S, Koner O, Menda F, Korkut K, Suzer K, Cakar N. The pulmonary and hemodynamic effects of two different recruitment maneuvers after cardiac surgery. Anesthesia and Analgesia 2007; 104(2): 384-90

64. Hedenstierna G, Edmark L. Mechanisms of atelectasis in the perioperative period. Best practice & research Clinical anaesthesiology 2010; 24(2): 157-69

65. Duggan M, Kavanagh BP. Pulmonary atelectasis: a pathogenic perioperative entity. Anesthesiology 2005; 102(4): 838-5466. Yoshino J, Akata T, Takahashi S. Intraoperative changes in arterial oxygenation during volume-controlled mechanical

ventilation in modestly obese patients undergoing laparotomies with general anesthesia. Acta Anaesthesiologica Scandinavica 2003; 47(6): 742-50

67. Valenza F, Vagginelli F, Tiby A, et al. Effects of the beach chair position, positive end-expiratory pressure, and pneumoperitoneum on respiratory function in morbidly obese patients during anesthesia and paralysis. Anesthesiology 2007; 107(5): 725-32

68. Hedenstierna G, Santesson J. Studies on intra-pulmonary gas distribution in the extremely obese. Influence of anaesthesia and artificial ventilation with and without positive end-expiratory pressure. Acta anaesthesiologica Scandinavica 1977; 21(4): 257-65

69. Perilli V, Sollazzi L, Modesti C, et al. Comparison of positive end-expiratory pressure with reverse Trendelenburg position in morbidly obese patients undergoing bariatric surgery: effects on hemodynamics and pulmonary gas exchange. Obesity surgery 2003; 13(4): 605-9

70. Gander S, Frascarolo P, Suter M, Spahn DR, Magnusson L. Positive end-expiratory pressure during induction of general anesthesia increases duration of nonhypoxic apnea in morbidly obese patients. Anesthesia and Analgesia 2005; 100(2): 580-4

71. Coussa M, Proietti S, Schnyder P, et al. Prevention of atelectasis formation during the induction of general anesthesia in morbidly obese patients. Anesthesia and Analgesia 2004; 98(5): 1491-5

72. Wetterslev J, Hansen EG, Roikjaer O, Kanstrup IL, Heslet L. Optimizing peroperative compliance with PEEP during upper abdominal surgery: effects on perioperative oxygenation and complications in patients without preoperative cardiopulmonary dysfunction. European journal of anaesthesiology 2001; 18(6): 358-65

73. Weingarten TN, Whalen FX, Warner DO, et al. Comparison of two ventilatory strategies in elderly patients undergoing major abdominal surgery. British journal of anaesthesia 2010; 104(1): 16-22

74. Wirth S, Baur M, Spaeth J, Guttmann J, Schumann S. Intraoperative positive end-expiratory pressure evaluation using the intratidal compliance-volume profile. British journal of anaesthesia 2015; 114(3): 483-90

75. Slutsky AS, Ranieri VM. Ventilator-induced lung injury. NEJM 2014; 370(10): 98076. Clark JM, Lambertsen CJ. Pulmonary oxygen toxicity: a review. Pharmacological reviews 1971; 23(2): 37-13377. Morse D, Otterbein LE, Watkins S, et al. Deficiency in the c-Jun NH2-terminal kinase signaling pathway confers

susceptibility to hyperoxic lung injury in mice. American journal of physiology Lung cellular and molecular physiology 2003; 285(1): L250-7

General introduction

Chap

ter

1

21

78. Crapo JD. Morphologic changes in pulmonary oxygen toxicity. Annual review of physiology 1986; 48: 721-3179. Crapo JD, Barry BE, Foscue HA, Shelburne J. Structural and biochemical changes in rat lungs occurring during exposures

to lethal and adaptive doses of oxygen. The American review of respiratory disease 1980; 122(1): 123-4380. Fracica PJ, Knapp MJ, Piantadosi CA, et al. Responses of baboons to prolonged hyperoxia: physiology and qualitative

pathology. Journal of applied physiology 1991; 71(6): 2352-6281. Kikuchi Y, Tateda K, Fuse ET, et al. Hyperoxia exaggerates bacterial dissemination and lethality in Pseudomonas

aeruginosa pneumonia. Pulmonary pharmacology & therapeutics 2009; 22(4): 333-982. Tateda K, Deng JC, Moore TA, et al. Hyperoxia mediates acute lung injury and increased lethality in murine Legionella

pneumonia: the role of apoptosis. Journal of immunology 2003; 170(8): 4209-1683. Li LF, Liao SK, Ko YS, Lee CH, Quinn DA. Hyperoxia increases ventilator-induced lung injury via mitogen-activated protein

kinases: a prospective, controlled animal experiment. Critical care 2007; 11(1): R2584. Makena PS, Luellen CL, Balazs L, et al. Preexposure to hyperoxia causes increased lung injury and epithelial apoptosis

in mice ventilated with high tidal volumes. American journal of physiology Lung cellular and molecular physiology 2010; 299(5): L711-9

85. Quinn DA, Moufarrej RK, Volokhov A, Hales CA. Interactions of lung stretch, hyperoxia, and MIP-2 production in ventilator-induced lung injury. Journal of applied physiology 2002; 93(2): 517-25.

86. Aggarwal NR, Brower RG. Targeting normoxemia in acute respiratory distress syndrome may cause worse short-term outcomes because of oxygen toxicity. Annals of the American Thoracic Society 2014; 11(9): 1449-53

87. Barazzone C, White CW. Mechanisms of cell injury and death in hyperoxia: role of cytokines and Bcl-2 family proteins. American journal of respiratory cell and molecular biology 2000; 22(5): 517-9

88. Edmark L, Auner U, Enlund M, Ostberg E, Hedenstierna G. Oxygen concentration and characteristics of progressive atelectasis formation during anaesthesia. Acta Anaesthesiologica Scandinavica 2011; 55(1): 75-81

89. de Jonge E, Peelen L, Keijzers PJ, et al. Association between administered oxygen, arterial partial oxygen pressure and mortality in mechanically ventilated intensive care unit patients. Critical Care 2008; 12(6): R156

90. Eastwood G, Bellomo R, Bailey M, et al. Arterial oxygen tension and mortality in mechanically ventilated patients. Intensive Care Medicine 2012; 38(1): 91-8

91. Aboab J, Jonson B, Kouatchet A, Taille S, Niklason L, Brochard L. Effect of inspired oxygen fraction on alveolar derecruitment in acute respiratory distress syndrome. Intensive Care Medicine 2006; 32(12): 1979-86

92. Kilgannon JH, Jones AE, Shapiro NI, et al. Association between arterial hyperoxia following resuscitation from cardiac arrest and in-hospital mortality. Jama 2010; 303(21): 2165-71

93. Brenner M, Stein D, Hu P, Kufera J, Wooford M, Scalea T. Association between early hyperoxia and worse outcomes after traumatic brain injury. Archives of surgery 2012; 147(11): 1042-6

94. Rincon F, Kang J, Maltenfort M, et al. Association between hyperoxia and mortality after stroke: a multicenter cohort study. Critical Care Medicine 2014; 42(2): 387-96

95. Bellomo R, Bailey M, Eastwood GM, et al. Arterial hyperoxia and in-hospital mortality after resuscitation from cardiac arrest. Critical care 2011; 15(2): R90

96. Young P, Beasley R, Bailey M, et al. The association between early arterial oxygenation and mortality in ventilated patients with acute ischaemic stroke. Critical care and resuscitation : journal of the Australasian Academy of Critical Care Medicine 2012; 14(1): 14-9

97. Raj R, Bendel S, Reinikainen M, et al. Hyperoxemia and long-term outcome after traumatic brain injury. Critical Care 2013; 17(4): R177

98. Damiani E, Adrario E, Girardis M, et al. Arterial hyperoxia and mortality in critically ill patients: a systematic review and metaanalysis. Critical care 2014; 18(6): 711

99. Helmerhorst HJ, Roos-Blom MJ, van Westerloo DJ, de Jonge E. Association Between Arterial Hyperoxia and Outcome in Subsets of Critical Illness: A Systematic Review, Metaanalysis, and Meta-Regression of Cohort Studies. Critical Care Medicine 2015

100. Wang CH, Chang WT, Huang CH, et al. The effect of hyperoxia on survival following adult cardiac arrest: a systematic review and metaanalysis of observational studies. Resuscitation 2014; 85(9): 1142-8

101. Farquhar H, Weatherall M, Wijesinghe M, et al. Systematic review of studies of the effect of hyperoxia on coronary blood flow. American heart journal 2009; 158(3): 371-7

102. Kenmure AC, Murdoch WR, Beattie AD, Marshall JC, Cameron AJ. Circulatory and metabolic effects of oxygen in myocardial infarction. British medical journal 1968; 4(5627): 360-4

103. McNulty PH, Robertson BJ, Tulli MA, et al. Effect of hyperoxia and vitamin C on coronary blood flow in patients with ischemic heart disease. Journal of applied physiology 2007; 102(5): 2040-5

104. Mak S, Azevedo ER, Liu PP, Newton GE. Effect of hyperoxia on left ventricular function and filling pressures in patients with and without congestive heart failure. Chest 2001; 120(2): 467-73

105. Stub D, Smith K, Bernard S, et al. Air Versus Oxygen in ST-Segment-Elevation Myocardial Infarction. Circulation 2015; 131(24): 2143-50

22

106. Patroniti N IG. Mechanical ventilation—Skills and techniques. Patient-centered Acute Care Training (PACT) Module—European Society of Intensive Care Medicine 2015

107. Edmark L, Kostova-Aherdan K, Enlund M, Hedenstierna G. Optimal oxygen concentration during induction of general anesthesia. Anesthesiology 2003; 98(1): 28-33

108. Hedenstierna G. Oxygen and anesthesia: what lung do we deliver to the post-operative ward? Acta Anaesthesiologica Scandinavica 2012; 56(6): 675-85

109. Benoit Z, Wicky S, Fischer JF, et al. The effect of increased FiO2 before tracheal extubation on postoperative atelectasis. Anesthesia and Analgesia 2002; 95(6): 1777-81, table of contents

110. Greif R, Laciny S, Rapf B, Hickle RS, Sessler DI. Supplemental oxygen reduces the incidence of postoperative nausea and vomiting. Anesthesiology 1999; 91(5): 1246-52

111. Orhan-Sungur M, Kranke P, Sessler D, Apfel CC. Does supplemental oxygen reduce postoperative nausea and vomiting? A metaanalysis of randomized controlled trials. Anesthesia and analgesia 2008; 106(6): 1733-8

112. Simurina T, Mraovic B, Mikulandra S, et al. Effects of high intraoperative inspired oxygen on postoperative nausea and vomiting in gynecologic laparoscopic surgery. Journal of clinical anesthesia 2010; 22(7): 492-8

113. Scifres CM, Leighton BL, Fogertey PJ, Macones GA, Stamilio DM. Supplemental oxygen for the prevention of postcesarean infectious morbidity: a randomized controlled trial. American journal of obstetrics and gynecology 2011; 205(3): 267 e1-9

114. Greif R, Akca O, Horn EP, Kurz A, Sessler DI. Supplemental perioperative oxygen to reduce the incidence of surgical-wound infection. NEJM 2000; 342(3): 161-7

115. Qadan M, Akca O, Mahid SS, Hornung CA, Polk HC, Jr. Perioperative supplemental oxygen therapy and surgical site infection: a metaanalysis of randomized controlled trials. Archives of surgery 2009; 144(4): 359-66

116. Pryor KO, Fahey TJ, 3rd, Lien CA, Goldstein PA. Surgical site infection and the routine use of perioperative hyperoxia in a general surgical population: a randomized controlled trial. JAMA 2004; 291(1): 79-87

117. Meyhoff CS, Wetterslev J, Jorgensen LN, et al. Effect of high perioperative oxygen fraction on surgical site infection and pulmonary complications after abdominal surgery: the PROXI randomized clinical trial. JAMA 2009; 302(14): 1543-50

118. Bustamante J, Tamayo E, Alvarez FJ, et al. Intraoperative PaO2 is not related to the development of surgical site infections after major cardiac surgery. Journal of cardiothoracic surgery 2011; 6: 4

119. Zoremba M, Dette F, Hunecke T, Braunecker S, Wulf H. The influence of perioperative oxygen concentration on postoperative lung function in moderately obese adults. European journal of anaesthesiology 2010; 27(6): 501-7

120. Hovaguimian F, Lysakowski C, Elia N, Tramer MR. Effect of intraoperative high inspired oxygen fraction on surgical site infection, postoperative nausea and vomiting, and pulmonary function: systematic review and metaanalysis of randomized controlled trials. Anesthesiology 2013; 119(2): 303-16

121. Wetterslev J, Meyhoff CS, Jorgensen LN, Gluud C, Lindschou J, Rasmussen LS. The effects of high perioperative inspiratory oxygen fraction for adult surgical patients. The Cochrane Database of Systematic Reviews 2015; 6: CD008884

122. Dongelmans DA, Veelo DP, Paulus F, et al. Weaning automation with adaptive support ventilation: a randomized controlled trial in cardiothoracic surgery patients. Anesthesia and Analgesia 2009; 108(2): 565-71

123. Dongelmans DA, Veelo DP, Binnekade JM, et al. Adaptive support ventilation with protocolized de-escalation and escalation does not accelerate tracheal extubation of patients after nonfast-track cardiothoracic surgery. Anesthesia and Analgesia 2010; 111(4): 961-7

General introduction

Chap

ter

1

23

Chapter 2

Intraoperative Ventilatory Strategies to Prevent Postoperative Pulmonary Complications – a Metaanalysis

Hemmes SNT, Serpa Neto A, Schultz MJ Current Opinion in Anesthesiology 2013; 26(2):126-33

26

Abstract

Purpose. It is uncertain whether patients undergoing short–lasting mechanical ventilation for surgery benefit from lung–protective intraoperative ventilatory settings including the use of lower tidal volumes, higher levels of positive end–expiratory pressure (PEEP) and/or recruitment manoeuvres (RM). We meta–analysed trials testing the effect of lung–protective intraoperative ventilatory settings on the incidence of postoperative pulmonary complications.

Recent findings. Eight articles (1669 patients) were included. Metaanalysis showed a decrease in lung injury development (risk ratio [RR] 0.40; 95% CI 0.22–0.70; I2 0%; number needed to treat [NNT] 37), pulmonary infection (RR 0.64; 95% CI 0.43–0.97; I2 0%; NNT 27) and atelectasis (RR 0.67; 95% CI 0.47–0.96; I2 48%; NNT 31) in patients receiving intraoperative MV with lower tidal volumes. Metaanalysis also showed a decrease in lung injury development (RR 0.29; 95% CI 0.14–0.60; I2 0%; NNT 29), pulmonary infection (RR 0.62; 95% CI 0.40–0.96; I2 15%; NNT 33) and atelectasis (RR 0.61; 95% CI 0.41–0.91; I2 0%; NNT 29) in patients ventilated with higher levels of PEEP, with or without RM.

Summary. Lung–protective intraoperative ventilatory settings may have the potential to protect against postoperative pulmonary complications.

Keywords. Mechanical ventilation, Intraoperative, Postoperative complications, Tidal volume, Positive end-expiratory pressure

Metaanalysis intraoperative ventilatory strategies

Chap

ter

2

27

Introduction

Mechanical ventilation (MV) has the potential to cause so–called ventilator–associated lung injury (VALI). VALI results from overdistention of non–dependent lung tissue causing excessive cyclic strain of alveolar cells,1 and repetitive opening and closing of dependent lung tissue resulting in cyclic cell stress due to the extreme forces exposed to lung cells at the interfaces between open and closed alveoli.2,3 Lung–protective MV with use of lower tidal volumes, which is suggested to prevent alveolar overdistention, benefits critically ill patients suffering from acute respiratory distress syndrome (ARDS).4 MV with higher levels of positive end–expiratory pressure (PEEP) with or without recruitment manoeuvres (RM), which is suggested to prevent repetitive opening and closing of alveoli, also seems beneficial at least in patients with severe ARDS.4 Recent clinical studies suggest MV with lower tidal volumes even to benefit critically ill patients without ARDS.5-7

MV is an essential supportive strategy during general anaesthesia for surgery. It is uncertain whether short–lasting MV during surgery also has the potential to cause VALI.8 However, both animal and human studies show VALI can develop shortly after initiation of MV.7,9,10 In addition, general anaesthesia causes large atelectasis, especially when muscle relaxants are used.11 As a consequence, there is an increased risk of overdistention of non–atelectatic lung tissue as well as repetitive opening and closing of partly atelectatic lung tissue. Thus, patients who need MV for surgery may also be vulnerable to the harmful effects of MV. Notably, surgical patients frequently suffer from postoperative pulmonary complications, with reported incidences of up to 5.0%.12,13 It is tempting to speculate on a causal relation between these complications and intraoperative ventilatory settings.

We hypothesize use of intraoperative lung–protective ventilatory settings to lower the incidence of postoperative pulmonary complications, and consequently on the postoperative clinical course and length of hospital stay. To test this hypothesis, we meta–analysed clinical trials of MV for surgery, focusing on the use of lower tidal volumes and/or higher levels of PEEP and RM. This is a secondary metaanalysis of a previously published metaanalysis of clinical trials testing lung–protective MV in patients who received short–term MV (i.e., in the operation room for surgery) or long–term MV (i.e., in the intensive care unit because of critical illness).14 The present metaanalysis is restricted to the clinical trials in the operation room.

Methods

We searched Medline (1966–2012), Cumulative Index to Nursing and Allied Health Literature (CINAHL), Web of Science, and Cochrane Central Register of Controlled Trials (CENTRAL). All reviewed articles and cross–referenced studies from retrieved articles were screened for pertinent information. Articles were selected for inclusion in the metaanalysis if they evaluated two types of MV in patients with uninjured lungs undergoing surgery. In one arm of the trial, MV should be protective (lower tidal volumes, and/or higher levels of PEEP with or without use of RMs). Then, this protective strategy should be compared with conventional methods

28

(higher tidal volumes, and no or lower levels of PEEP and no use of RMs) in the other arm of the trial. We excluded trials of patients undergoing cardiac surgery. We also excluded revisions and trials that did not report the outcomes of interest (defined below). When we found duplicate articles of the same trial in preliminary abstracts and articles, we analysed data from the most complete data set.

Data were extracted from each article using a data recording form developed for the previously published metaanalysis.14 After extraction, data were reviewed and compared by the second author. Whenever needed, we obtained additional information about a specific study by directly questioning the principal investigator of the specific trial.

The primary endpoint was the incidence of lung injury in each arm of the trial. Secondary endpoints included incidence of pulmonary infection (using the authors’ definition) or atelectasis. Statistical analysis was performed as described in the original metaanalysis.14

Results

The initial search yielded 2.123 articles (459 from MEDLINE, 141 from CENTRAL, 885 from CINAHL, and 638 from Web of Science) (figure 1). After removing 711 duplicate articles, we evaluated the abstracts of 1.412 articles. After evaluating them, 1.364 articles were excluded because they did not meet inclusion criteria. Another five articles were excluded because MV was applied for other reasons than surgery, no data on outcome of interest was reported in 28, and same cohort previously analysed in seven. Finally, eight articles were included in the final analysis.15-22

Tidal volume reductionOur search of the literature revealed eight articles (1669 patients) reporting on trials comparing lower with conventional tidal volumes during surgery (table 1 and table 2). Metaanalysis of these trials showed that 17 of 858 patients (2.0%) ventilated with lower tidal volumes and 36 of 755 patients (4.7%) ventilated with conventional tidal volumes developed lung injury during follow-up (risk ratio [RR] 0.40; 95% confidence interval [CI] 0.22–0.70; number needed to treat [NNT] 37) (figure 2). The analysis displayed no signs of heterogeneity (I2 = 0%). Pulmonary infection and atelectasis showed lower incidence in patients receiving lower tidal volume ventilation (RR 0.64; 95% CI 0.43–0.97; NNT 27 and RR 0.67; 95% CI 0.47–0.96; NNT 31, respectively). The I2 test indicated no heterogeneity in the analysis of pulmonary infection, but moderate heterogeneity in the analysis of atelectasis (0% and 48% respectively).

Higher levels of PEEP and RMsOur search of the literature revealed five articles (1323 patients) reporting on trials comparing no or lower levels of PEEP (up to 3 cmH2O) with higher levels of PEEP (from 3 to 12 cmH2O) during surgery (table 1).

Metaanalysis of these trials shows that 9 of 654 patients (1.4%) ventilated with higher levels of PEEP developed postoperative lung injury compared to 31 of 629 patients (4.9%) receiving

Metaanalysis intraoperative ventilatory strategies

Chap

ter

2

29

lower levels of PEEP (RR 0.29; 95% CI 0.14–0.60; NNT 29) (figure 3), without any signs for heterogeneity within the analysis (I2 = 0%). A beneficial effect of higher levels of intraoperative PEEP on postoperative pulmonary infection and atelectasis was also found (RR 0.62; 95% CI 0.40–0.96; NNT 33 and RR 0.61; 95% CI 0.41–0.91; NNT 29, respectively). The I2 test indicated moderate heterogeneity in the analysis of pulmonary infection, but not in the analysis of atelectasis (15% and 0% respectively). We did not find trials specifically investigating exclusively the effects of intraoperative RM.

Figure 1. Literature search strategy; ARDS indicates acute respiratory distress syndrome

30

Tabl

e 1.

Cha

ract

eris

tics

of th

e in

clud

ed st

udie

s an

d su

mm

ary

of c

ontin

uous

var

iabl

es

Prot

ectiv

eCo

nser

vativ

ePr

otec

tive

Cons

erva

tive

Stud

yN

VTPE

EPRM

NVT

PEEP

RMN

Setti

ngD

esig

nFU

Tim

e of

MV

Prim

ary

Out

com

e

Mic

hele

t1552

55

N26

90

N26

OS

RCT

187.

06 ±

1.8

17.

76 ±

1.8

5Cy

toki

nes i

n bl

ood

Cai16

166

0N

810

0N

8N

euro

RCT

7.15

6.90

± 2

.20

7.4

± 3.

10CT

Ate

lect

asis

Lin7

405

3-5

N20

90

N20

OS

RCT

244.

33 ±

0.9

04.

23 ±

0.7

1Cy

toki

nes i

n bl

ood

Lick

er18

1091

63

Y55

89

3N

533

OS

COH

---2.

93 ±

1.2

02.

76 ±

1.0

LI

Wei

ngar

ten19

406

12Y

2010

0N

20Su

rgic

alRC

TDi

scha

rge

5.13

± 1

.86

5.73

± 1

.71

Oxy

gena

tion

Bust

aman

te20

229

84

N15

410

4N

75Su

rgic

alCR

O---

NS

NS

TMV;

ICU

LS;

Mor

talit

y

Yang

2110

06

5N

5010

0N

50O

SRC

T16

82.

00 ±

0.6

82.

11 ±

0.8

0LI

Tres

chan

2210

16

5Y

5012

5Y

51Su

rgic

alRC

T12

08.

70 ±

5.2

08.

70 ±

5.9

0Sp

irom

etry

Tota

l1,

669

6.14

±

0.86

4.50

(3

.0

-5.0

)---

886

10.3

5 ±

1.15

0 (0

- 3.

75)

---78

3---

---6.

57 (4

.50

– 19

.50)

6.90

(3.6

3 –

8.70

)7.

40 (3

.49

– 10

.35)

---

Mea

n ±

stan

dard

dev

iatio

n; M

edia

n (in

terq

uarti

le ra

nge)

; VT:

Tid

al v

olum

e (in

mL/

kg);

FU: F

ollo

w-u

p; O

S: O

ncol

ogy

surg

ery;

RCT

: Ran

dom

ized

cont

rolle

d tr

ial;

COH:

Coh

ort;

CRO

: Cr

oss-

secti

onal

; CT:

Com

pute

d to

mog

raph

y; N

S: N

ot sp

ecifi

ed; L

I: Lu

ng in

jury

; TM

V: T

ime

of m

echa

nica

l ven

tilati

on; I

CULS

: ICU

leng

th o

f sta

y

Metaanalysis intraoperative ventilatory strategies

Chap

ter

2

31

Figure 2. Effect of intraoperative ventilation with lower tidal volumes

32

Table 2. Synthesis of demographic, ventilatory and laboratorial characteristics of the patients in the final follow-up

Protective Ventilation (n = 886)

Conventional Ventilation (n = 73) p-value

Age, years 60.27 ± 8.31 60.33 ± 8.06 0.910

Weight, kg 73.04 ± 13.04 73.01 ± 12.56 0.965

Tidal volume, ml/kg IBWa 6.14 ± 0.86 10.35 ± 1.15 < 0.0001

PEEP, cmH2Oa 6.62 ± 2.65 2.74 ± 2.82 0.001

Plateau pressure, cmH2Oa 16.62 ± 2.76 20.45 ± 2.54 0.021

Respiratory rate, beats per minutea 16.62 ± 2.72 10.78 ± 2.67 0.007

Minute-ventilation, litres/minutea 7.76 ± 2.61 8.56 ± 2.58 0.917

PaO2 / FiO2a 332.86 ± 61.48 339.68 ± 67.70 0.797

PaCO2, mmHga 41.86 ± 3.32 39.05 ± 3.42 0.052

pHa 7.35 ± 0.03 7.39 ± 0.03 0.073

Mean ± standard deviation; IBW: ideal body weight; PEEP: positive end expiratory pressure; a: in the final of the follow-up

Discussion

This metaanalysis suggests that intraoperative MV with lower tidal volumes may protect surgical patients from development of postoperative lung injury, pulmonary infections and atelectasis. This metaanalysis also suggests that intraoperative use of higher levels of PEEP during MV attenuates development of lung injury, pulmonary infection and atelectasis.

Implementation of lung–protective MV for surgery has the potential to significantly reduce postoperative pulmonary complications. Considering the high number of surgical procedures performed worldwide daily,23 reduction of postoperative pulmonary complications could be of great importance. Notably, a recent international prospective trial shows the incidence of postoperative mortality to be as high as 4%, much higher than previously assumed.24 A large international observational study is underway to address the effect of intraoperative ventilatory settings on postoperative complications.25

Prescription of MV in critically ill patients has definitely changed over the last decades. There has been progressive reduction of tidal volume size, from > 12 ml/kg in the 1970s 26,27 to < 9 ml/kg in more recent epidemiologic studies of MV practice in Europe and the Americas.28-31 This change was largely stimulated by results from animal studies, which clearly show injurious tidal volume settings to aggravate pre–existing pulmonary injury.9 Several clinical trials confirm the existence of VALI by showing reduced morbidity and mortality in patients with ARDS ventilated with lower tidal volumes.4 While initially intensive care unit physicians have been reluctant to use lower tidal volumes as part of their MV strategy, guidelines now strongly support the use

Metaanalysis intraoperative ventilatory strategies

Chap

ter

2

33

of lower tidal volumes in patients with ARDS, e.g., in patients with sepsis.32 Critically ill patients without ARDS also seem to benefit from ventilation with lower tidal volumes.5,6 One recent randomized controlled trial shows a lower tidal volume strategy to protect against lung injury in patients without ARDS at onset of MV in the intensive care unit.7

Notably, a recent observational study in patients undergoing short–term postoperative MV after cardiac surgery shows MV with tidal volumes > 10 ml/kg to be associated with prolonged MV, hemodynamic instability, multiple organ failure, and prolonged stay in the ICU, compared to MV with lower tidal volumes. In this study women and obese patients are found to be particularly at risk of receiving ventilation with too large tidal volumes.33 These results confirm, at least in part findings from another recent study that identifies female gender, overweight and underweight as independent factors for MV with too large tidal volumes.34

MV with lower tidal volumes may not come without challenges. Use of lower tidal volumes could increase cyclic alveolar collapse of dependent lung regions, raising the risk of atelectrauma. Application of PEEP is an easy intervention that may counteract this side–effect of lower tidal volume ventilation. Lower tidal volume ventilation could also lead to hypercapnia and hypercapnic acidosis. Notably, so-called permissive hypercapnia is thought to have lung–protective qualities,

Figure 3. Effect of intraoperative ventilation with higher levels of PEEP

34

even though the exact clinical implications are not entirely clear.35

Prescription of PEEP in critically ill patients has also changed over the last decades. PEEP is progressively more frequent applied in intubated and mechanically ventilated patients in the intensive care unit, with an increase in use of PEEP levels > 10 cmH2O from 28% in the late ‘90s 28,29 to 40% in a more recent survey across ICUs in Europe and the Americas.36 Particularly in patients with ARDS higher levels of PEEP are being applied, even though the benefits of higher PEEP levels with or without RM are not unequivocally demonstrated.28,29,36 Use of higher levels of PEEP and RM could benefit patients with severe ARDS, though .4,37 Trials investigating the effects of higher levels of PEEP in critically ill patients without ARDS are lacking. The results of the present metaanalysis are in line with results from a previous systematic review suggesting higher levels of PEEP to reduce postoperative atelectasis.38

None of the trials included in our metaanalysis analysed the effect of RM separate from the use of higher levels of PEEP. However, one recent randomized controlled trial of cardiac surgery patients shows decrease of alveolar dead–space and increase in arterial oxygenation during surgery when RMs are performed.39

An adversity of use of higher levels of PEEP, with or without the use of RM, may lay in an increase in right ventricular afterload as well as a decrease in right ventricular preload. This could cause a decrease in left ventricular preload and reduction in left ventricular stroke volume.40 It is uncertain whether this causes problems in patients undergoing surgery.

Our study knows several shortcomings. First, it is difficult if not impossible to differentiate between the beneficial effect from lower tidal volumes and that from higher levels of PEEP with or without RM. Most trials included in this metaanalyses compared “conventional ventilation” with higher tidal volumes and low levels of PEEP with a “lung-protective” MV strategy with lower tidal volume ventilation and higher levels of PEEP (table 1). As a result all trials included in the metaanalysis assessing the effect of higher levels of PEEP and RM is also part of the metaanalysis analysing the effect of lower tidal volumes. Second, the incidence of lung injury could very well be higher than calculated in this metaanalysis, as the clinical picture of ARDS often resembles “suspected” pulmonary infection. ARDS as well as pulmonary infection may present with leucocytosis, fever and pulmonary infiltrates on the chest radiograph. Thus, it could be that ARDS is mistakenly diagnosed as pulmonary infection in some cases. Finally, when interpreting the results of this metaanalysis, the possible occurrence of positive publication bias should be taken into account. Furthermore the use of funnel plots has a limited role, as test for bias when the number of studies included in the analysis is small. Despite these limitations, our metaanalysis provides interesting information which needs further exploration.

Metaanalysis intraoperative ventilatory strategies

Chap

ter

2

35

Conclusion

Lower tidal volumes, higher levels of PEEP and RM are increasingly used in intensive care unit patients receiving long–term MV. Intraoperative use of lower tidal volumes could reduce the incidence of postoperative lung injury, pulmonary infections and atelectasis. Intraoperative use of higher levels of PEEP and RMs also reduce the incidence of these complications. It is difficult if not impossible to separate the beneficial effects of lower tidal volumes from that of higher levels of PEEP and RM. To better establish the effect of lung–protective intraoperative ventilatory settings we are also in need of well–powered randomized clinical trials. Presently, one large multicentre trial is conducted to identify the effect of intraoperative use of higher levels of PEEP and RM on the incidence of postoperative complications in adult surgical patients.41

Key points

1. Intraoperative use of lower tidal volumes may reduce postoperative lung injury, pulmonary infections and atelectasis

2. It is uncertain whether higher levels of PEEP, with or without use of RM, adds to the beneficial effects of intraoperative use of lower tidal volumes

3. We are in need of well–powered randomized controlled trials that test the effect of intraoperative lung–protective ventilatory settings, including tidal volume size, higher levels of PEEP and RM.

36

References1. Slutsky AS. Lung injury caused by mechanical ventilation. Chest 1999; 116: 9S-15S2. Mead J, Takishima T, Leith D. Stress distribution in lungs: a model of pulmonary elasticity. J Appl Physiol 1970; 28:

596-6083. Gattinoni L, Carlesso E, Caironi P. Stress and strain within the lung. Curr Opin Crit Care 2012; 18: 42-74. Putensen C, Theuerkauf N, Zinserling J, et al. Metaanalysis: ventilation strategies and outcomes of the acute respiratory

distress syndrome and acute lung injury. Ann Intern Med 2009; 151: 566-765. Gajic O, Dara SI, Mendez JL, et al. Ventilator-associated lung injury in patients without acute lung injury at the onset

of mechanical ventilation. Crit Care Med 2004; 32: 1817-246. Gajic O, Frutos-Vivar F, Esteban A, et al. Ventilator settings as a risk factor for acute respiratory distress syndrome in

mechanically ventilated patients. Intensive Care Med 2005; 31: 922-67. Determann RM, Royakkers A, Wolthuis EK, et al. Ventilation with lower tidal volumes as compared with conventional

tidal volumes for patients without acute lung injury: a preventive randomized controlled trial. Crit Care 2010; 14: R18. Schultz MJ, Haitsma JJ, Slutsky AS, et al. What tidal volumes should be used in patients without acute lung injury?

Anesthesiology 2007; 106: 1226-319. Dreyfuss D, Saumon G. Ventilator-induced lung injury: lessons from experimental studies. Am J Respir Crit Care Med

1998; 157: 294-32310. Wolthuis EK, Vlaar AP, Choi G, et al. Mechanical ventilation using non-injurious ventilation settings causes lung injury

in the absence of pre-existing lung injury in healthy mice. Crit Care 2009; 13: R111. Duggan M, Kavanagh BP. Pulmonary atelectasis: a pathogenic perioperative entity. Anesthesiology 2005; 102: 838-5412. Smetana GW, Lawrence VA, Cornell JE. Preoperative pulmonary risk stratification for noncardiothoracic surgery:

systematic review for the American College of Physicians. Ann Intern Med 2006; 144: 581-9513. Canet J, Gallart L, Gomar C, et al. Prediction of Postoperative Pulmonary Complications in a Population-based Surgical

Cohort. Anesthesiology 2010; 113: 1338-5014. Serpa Neto A, Cardoso SO, Manetta JA, et al. Association between use of lung-protective ventilation with lower tidal

volumes and clinical outcomes among patients without acute respiratory distress syndrome: a metaanalysis. JAMA 2012; 308: 1651-9

15. Michelet P, D’Journo XB, Roch A, et al. Protective ventilation influences systemic inflammation after esophagectomy: a randomized controlled study. Anesthesiology 2006; 105: 911-9

16. Cai H, Gong H, Zhang L, et al. Effect of low tidal volume ventilation on atelectasis in patients during general anesthesia: a computed tomographic scan. J Clin Anesth 2007; 19: 125-9

17. Lin WQ, Lu XY, Cao LH, et al. [Effects of the lung protective ventilatory strategy on proinflammatory cytokine release during one-lung ventilation]. Ai Zheng 2008; 27: 870-3

18. Licker M, Diaper J, Villiger Y, et al. Impact of intraoperative lung-protective interventions in patients undergoing lung cancer surgery. Crit Care 2009; 13: R41

19. Weingarten TN, Whalen FX, Warner DO, et al. Comparison of two ventilatory strategies in elderly patients undergoing major abdominal surgery. Br J Anaesth 2010; 104: 16-22

20. Fernandez-Bustamante A, Wood CL, Tran ZV, et al. Intraoperative ventilation: incidence and risk factors for receiving large tidal volumes during general anesthesia. BMC Anesthesiol 2011; 11: 22

21. Yang M, Ahn HJ, Kim K, et al. Does a protective ventilation strategy reduce the risk of pulmonary complications after lung cancer surgery?: a randomized controlled trial. Chest 2011; 139: 530-7

22. Treschan TA, Kaisers W, Schaefer MS, et al. Ventilation with low tidal volumes during upper abdominal surgery does not improve postoperative lung function. Br J Anaesth 2012; 109: 263-71

23. Weiser TG, Regenbogen SE, Thompson KD, et al. An estimation of the global volume of surgery: a modelling strategy based on available data. Lancet 2008; 372: 139-44

24. Pearse RM, Moreno RP, Bauer P, et al. Mortality after surgery in Europe: a 7 day cohort study. Lancet 2012 380: 1059-6525. ClinicalTrials.gov. (2012) Local Assessment of Ventilatory Management During General Anesthesia for Surgery (LAS

VEGAS), NCT01601223. http://clinicaltrials.gov/ct2/show/NCT01601223?term=las+vegas&rank=1 26. Suter PM, Fairley B, Isenberg MD. Optimum end-expiratory airway pressure in patients with acute pulmonary failure.

NEJM 1975; 292: 284-927. Jardin F, Farcot JC, Boisante L, et al. Influence of positive end-expiratory pressure on left ventricular performance.

NEJM 1981; 304: 387-9228. Esteban A, Anzueto A, Alia I, et al. How is mechanical ventilation employed in the intensive care unit? An international

utilization review. Am J Respir Crit Care Med 2000; 161: 1450-829. Esteban A, Anzueto A, Frutos F, et al. Characteristics and outcomes in adult patients receiving mechanical ventilation:

a 28-day international study. JAMA 2002; 287: 345-55

Metaanalysis intraoperative ventilatory strategies

Chap

ter

2

37

30. Brun-Buisson C, Minelli C, Bertolini G, et al. Epidemiology and outcome of acute lung injury in European intensive care units. Results from the ALIVE study. Intensive Care Med 2004; 30: 51-61

31. Sakr Y, Vincent JL, Reinhart K, et al. High tidal volume and positive fluid balance are associated with worse outcome in acute lung injury. Chest 2005; 128: 3098-108

32. Dellinger RP, Levy MM, Carlet JM, et al. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock: 2008. Crit Care Med 2008; 36: 296-327

33. Lellouche F, Dionne S, Simard S, et al. High tidal volumes in mechanically ventilated patients increase organ dysfunction after cardiac surgery. Anesthesiology 2012; 116: 1072-82

34. Jaber S, Coisel Y, Chanques G, et al. A multicentre observational study of intra-operative ventilatory management during general anaesthesia: tidal volumes and relation to body weight. Anaesthesia 2012; 67: 999-1008

35. Ismaiel NM, Henzler D. Effects of hypercapnia and hypercapnic acidosis on attenuation of ventilator-associated lung injury. Minerva Anestesiol 2011; 77: 723-33.

36. Esteban A, Ferguson ND, Meade MO, et al. Evolution of mechanical ventilation in response to clinical research. Am J Respir Crit Care Med 2008; 177: 170-7

37. Gattinoni L, Caironi P. Refining ventilatory treatment for acute lung injury and acute respiratory distress syndrome. JAMA 2008; 299: 691-3

38. Imberger G, McIlroy D, Pace NL, et al. Positive end-expiratory pressure (PEEP) during anaesthesia for the prevention of mortality and postoperative pulmonary complications. Cochrane Database Syst Rev 2010: CD007922

39. Unzueta C, Tusman G, Suarez-Sipmann F, et al. Alveolar recruitment improves ventilation during thoracic surgery: a randomized controlled trial. Br J Anaesth 2012; 108: 517-24

40. Luecke T, Pelosi P. Clinical review: Positive end-expiratory pressure and cardiac output. Crit Care 2005; 9: 607-2141. Hemmes SN, Severgnini P, Jaber S, et al. Rationale and study design of PROVHILO - a worldwide multicenter randomized

controlled trial on protective ventilation during general anesthesia for open abdominal surgery. Trials 2011; 12: 111

Chapter 3

Intraoperative Protective Mechanical Ventilation for Prevention of Postoperative Pulmonary ComplicationsA Comprehensive Review of the Role of Tidal Volume, Positive End-Expiratory Pressure and Lung Recruitment Manoeuvres

Güldner A, Kiss T, Serpa Neto A, Hemmes SNT, Canet J, Spieth PM, Rocco PRM, Schultz MJ, Pelosi P, Gama de Abreu M. Anesthesiology 2015; 123(3): 692-713

40

Abstract

Postoperative pulmonary complications are associated with increased morbidity, length of hospital stay, and mortality following major surgery. Intraoperative lung-protective mechanical ventilation has the potential to reduce the incidence of postoperative pulmonary complications. This review discusses the relevant literature on definition of and methods to predict occurrence of postoperative pulmonary complication, the pathophysiology of ventilator-induced lung injury with emphasis to the non-injured lung, and protective ventilation strategies, including the respective roles of tidal volumes, positive end-expiratory pressure and recruitment manoeuvres. The authors propose an algorithm for protective intraoperative mechanical ventilation based upon evidence from recent randomized clinical trials.

Review intraoperative protective ventilation

Chap

ter

3

41

Introduction

Postoperative pulmonary complications (PPCs) can have an important impact on the morbidity and mortality of patients who need major surgery.1 Approximately 5% of patients undergoing general surgery will develop a PPC and one of five patients who developed a PPC die within 30 days of surgery.1 Furthermore, the number of PPCs is strongly associated with postoperative length of stay and short-term and long-term mortality.1,2

There is growing evidence that intraoperative lung-protective mechanical ventilation using low tidal volumes, with or without high levels of positive end–expiratory pressure (PEEP) and recruitment manoeuvres, prevents PPCs compared to mechanical ventilation with high tidal volumes and low levels of PEEP without recruitment maneuvers.3-6

In the present article, we review the definition of and methods to predict PPCs, the patho-physiology of ventilator-induced lung injury (VILI) with emphasis on the non-injured lung, and ventilation strategies to minimize PPCs. To identify the most recent evidence from the literature on randomized clinical trials (RCTs) addressing intraoperative mechanical ventilation and non-clinical as well as clinical postoperative outcome measures, we conducted a MEDLINE review using the following search terms: (‘lower tidal volume’ OR ‘low tidal volume’ OR ‘protective ventilation’ OR ‘recruitment manoeuvres’ OR ‘PEEP’ OR ‘positive end expiratory pressure’). Retrieved articles, and cross–referenced studies from those articles, were screened for pertinent information.

Definition and prediction of postoperative pulmonary complications Summary of current definitionsPPCs are usually presented as a composite, which then includes possible fatal and non-fatal respiratory events of new onset occurring in the postoperative period. Currently, there is no agreement about which of these events should be considered as PPC, for example respiratory failure, lung injury, pneumonia, prolonged or unplanned mechanical ventilation or intubation, hypoxemia, atelectasis, bronchospasm, pleural effusion, pneumothorax, ventilatory depression, and aspiration pneumonitis.7,8 From a clinical standpoint, it is worthwhile to present PPCs as a composite, because any of these events alone or their associations has a significant impact on the postoperative outcome,1 using different definitions. However, it is clear that these events can have different pathophysiologic mechanisms. For this reason, some studies have focused on single events, mainly respiratory failure9 and pneumonia.10

PPCs, to be considered as such, must be related to anesthesia and/or surgery. Furthermore, the time frame must be well defined. Usually, an event is only considered as PPC if it develops within 5 to 7 days after surgery.8,11

42

Prediction of Postoperative Pulmonary ComplicationsPrediction of PPCs, or any of the single postoperative respiratory events that is part of that composite, can be useful to plan perioperative strategies aiming at their prevention, and also to reduce health system costs.12 First, the risk factors associated with the development of PPCs must be identified. In 2006, the American College of Physicians published a systematic review of the literature listing a number of risk factors for PPCs according to their respective levels of evidence.13 In recent years, that list has been expanded to include other factors found to increase the risk of PPCs. Table 1 depicts risk factors associated with PPCs according to the current literature. Approximately 50% of the risk for PPCs are attributable to the patient’s health conditions, while the other 50% are related to the surgical procedure and the anaesthetic management itself.1

Table 1. Risk factors for postoperative pulmonary complications

Patient characteristics Preoperative testing Surgery Anaesthetic management

Age Low albumin Open thoracic surgery General anesthesia

Male sex Low SpO2 (≤ 95%) Cardiac surgery High respiratory driving pressure (≥ 13 cmH2O)

ASA class ≥ 3 Anaemia (Hb < 10 g/dL) Open upper abdominal surgery High inspiratory oxygen fraction

Previous respiratory infection

Major vascular surgery

High volume of crystalloid administration

Functional dependency Neurosurgery Red blood cell transfusion

Congestive heart failure Urology Residual neuromuscular blockade

COPD Duration of surgery > 2h Nasogastric tube use

Smoking Emergent surgery

Renal failure

Gastroesophageal reflux disease

Weight loss

ASA, American Society of Anesthesiologists; COPD, chronic obstructive pulmonary disease; Hb, haemoglobin concentration; PPCs, postoperative pulmonary complications; SpO2, oxygen saturation as measured by pulse oximetry. Respiratory driving pressure, defined as inspiratory plateau airway pressure minus positive end-expiratory pressure

Based on risk factors, different scores have been developed that have the potential to predict the occurrence of PPCs,6,14-16 as shown in table 2. However, their applicability may be limited since they were derived from restricted settings,16 retrospective databases,15 or only validated for specific PPCs.6,14 The Assess Respiratory RIsk in Surgical Patients in CATalonia (ARISCAT) study was conducted in a general surgical population of Catalonia, Spain.1 After a multivariate analysis, a score based on seven risk factors was developed and underwent internal validation, showing

Review intraoperative protective ventilation

Chap

ter

3

43

a clinically relevant predictive capability (c-statistic, 0.90). Recently, the ARISCAT score was externally validated in a large European surgical sample (the Prospective Evaluation of a RIsk Score for Postoperative Pulmonary COmPlications in Europe, PERISCOPE study).2 Although differences in the performance of the ARISCAT score have been observed between European geographic areas, the score was able to discriminate three levels of PPCs risk (low, intermediate and high). Thus, at present, the ARISCAT score may represent the most valuable tool for predicting PPCs across different countries and surgical populations.

Putative mechanisms of ventilator-induced lung injury

The coexistence of closed, recruitable and already overdistended alveolar regions makes the lung vulnerable to detrimental effects of mechanical stress and strain induced by mechanical ventilation.17,18 The physical forces in some alveolar regions may exceed the elastic properties of the lungs although gross measurements of airway pressures or lung mechanics as usually monitored under anesthesia still suggest mechanical ventilation is in a “safe” zone.19,20 Several mechanisms have been postulated to describe the development of VILI.21 Increased airway pressure (barotrauma) or the application of high tidal volumes (volutrauma) may cause damage or disruption of alveolar epithelial cells, by generating transpulmonary pressures (stress) that exceed the elastic properties of the lung parenchyma above its resting volume (strain).22,23 It has been demonstrated that the duration of mechanical stress defined as the stress versus time product affects the development of pulmonary inflammatory response.24 While high stress versus time product increased the gene expression of biological markers associated with inflammation and alveolar epithelial cell injury and low stress versus time product increased the molecular markers of endothelial cell damage, balanced stress versus time product as defined by an inspiratory to expiratory time ratio of 1:1 was associated with attenuated lung damage.24 Especially in the presence of atelectasis, mechanical ventilation may cause damage by repetitive collapse and reopening of alveolar units, a phenomenon known as atelectrauma.25 All three mechanisms, namely barotrauma, volutrauma and atelectrauma may affect alveolar as well as vascular epithelial and endothelial cells26,27 as well as promote extracellular matrix fragmentation.28,29

The extracellular matrix of the lung parenchyma seems to be particularly sensitive to stress from mechanical ventilation, as illustrated in figure 1. Initially, the proteoglycans on the endothelial side and between the endothelial and epithelial lines undergo damage dependent on tidal volume,28 as well as breathing pattern.29 The mechanical fragmentation of the extracellular matrix promotes interstitial oedema and activation of metalloproteinases, further damaging the extracellular matrix itself. In a second step, fragments of the extracellular matrix can promote activation of inflammatory mediators.30,31 Furthermore, the damage of the extracellular matrix induced by mechanical ventilation might be exacerbated by fluid load,32 which is not uncommon during general anesthesia. However, fluid overload seems to minimize the inflammatory response, likely by dilution of extracellular matrix fragments or changes in their structure, thereby down regulating the local inflammatory response.32 This suggests that: 1) injurious mechanical ventilation makes the lung more susceptible to further insults; 2) in previously healthy lungs, VILI can be induced without early increase in inflammatory mediators.

44

Tabl

e 2.

Sco

res

for p

redi

ction

of p

osto

pera

tive

pulm

onar

y co

mpl

icati

ons

Refe

renc

e/Ye

ar

publ

ishe

dSt

udy

desi

gnPa

tient

pop

ulati

onN

umbe

r of

patie

nts

Scor

e ac

rony

mSc

orin

g sy

stem

Cut-

offQ

ualit

y of

pr

edic

tion

Pred

ictio

n of

gen

eral

pos

tope

rativ

e pu

lmon

ary

com

plic

ation

s

Cane

t et.

al.,

2010

1Pr

ospe

ctive

, m

ultic

ente

r, ob

serv

ation

al

coho

rt st

udy

Adul

t pati

ents

un

derg

oing

no

nobs

tetr

ic

surg

ical

pr

oced

ures

un

der g

ener

al,

neur

axia

l, or

regi

onal

an

esth

esia

2,46

4 ov

eral

l1,

624

deriv

ation

837

valid

ation

3 pa

tient

s with

m

issin

g da

ta fo

r tw

o pa

ram

eter

s re

leva

nt fo

r th

e sc

ore

(SpO

2+re

spira

tory

in

fecti

on d

urin

g la

st m

onth

)

ARIS

CAT

Asse

ss

Resp

irato

ry

Risk

in S

urgi

cal

Patie

nts i

n Ca

talo

nia

Age

51–8

0 >8

0Sp

O2%

91

–95

<9

0Re

spira

tory

infe

ction

<3

0 da

ysPr

eope

rativ

e an

aem

ia (H

b <1

0 g/

dl)

Surg

ical

inci

sion:

Perip

hera

lU

pper

abd

omin

alIn

trat

hora

cic

Dura

tion

of su

rger

y:

≤2h

>2-3

>3

Emer

genc

y pr

oced

ure

3 16 8 24 17 11 1 15 24 1 16 23 8

Leve

l/Po

int/

Rate

of P

PC

low

/<26

/1.6

%;

med

ium

/26-

44/ 1

3.3%

high

/≥45

/42.

1%(v

alid

ation

subs

ampl

e)

PPC:

Resp

irato

ry in

fecti

on,

resp

irato

ry fa

ilure

, ple

ural

eff

usio

n, a

tele

ctas

is,

pneu

mot

hora

x,

bron

chos

pasm

, asp

iratio

n pn

eum

oniti

s

Deriv

ation

co

hort

AUC:

0.8

9

Valid

ation

co

hort

AUC:

0.8

4

Maz

o et

. al.

2014

2

(Val

idati

on

of A

RISC

AT

scor

e fr

om

Cane

t et.

al.,

2010

1 in

a la

rger

, in

tern

ation

al,

mul

ticen

ter

coho

rt)

Pros

pecti

ve,

mul

ticen

ter,

obse

rvati

onal

co

hort

stud

y

Adul

t pati

ents

un

derg

oing

no

nobs

tetr

ic

surg

ical

pr

oced

ures

un

der g

ener

al,

neur

axia

l, or

pl

exus

blo

ck

anes

thes

ia

5,09

9AR

ISCA

TAs

sess

Re

spira

tory

Ri

sk in

Sur

gica

l Pa

tient

s in

Cata

loni

a

see

abov

eLe

vel/

Poin

t/Pr

edic

ted/

O

bser

ved

Rate

of P

PC

low

/<26

/0.8

7%/ 3

.39%

med

ium

/26-

44/

7.82

%/1

2.96

%hi

gh/≥

45/3

8.13

%/ 3

8.01

%

AUC:

0.8

0

Review intraoperative protective ventilation

Chap

ter

3

45

Tabl

e 2.

Sco

res

for p

redi

ction

of p

osto

pera

tive

pulm

onar

y co

mpl

icati

ons

Refe

renc

e/Ye

ar

publ

ishe

dSt

udy

desi

gnPa

tient

pop

ulati

onN

umbe

r of

patie

nts

Scor

e ac

rony

mSc

orin

g sy

stem

Cut-

offQ

ualit

y of

pr

edic

tion

Pred

ictio

n of

gen

eral

pos

tope

rativ

e pu

lmon

ary

com

plic

ation

s

Cane

t et.

al.,

2010

1Pr

ospe

ctive

, m

ultic

ente

r, ob

serv

ation

al

coho

rt st

udy

Adul

t pati

ents

un

derg

oing

no

nobs

tetr

ic

surg

ical

pr

oced

ures

un

der g

ener

al,

neur

axia

l, or

regi

onal

an

esth

esia

2,46

4 ov

eral

l1,

624

deriv

ation

837

valid

ation

3 pa

tient

s with

m

issin

g da

ta fo

r tw

o pa

ram

eter

s re

leva

nt fo

r th

e sc

ore

(SpO

2+re

spira

tory

in

fecti

on d

urin

g la

st m

onth

)

ARIS

CAT

Asse

ss

Resp

irato

ry

Risk

in S

urgi

cal

Patie

nts i

n Ca

talo

nia

Age

51–8

0 >8

0Sp

O2%

91

–95

<9

0Re

spira

tory

infe

ction

<3

0 da

ysPr

eope

rativ

e an

aem

ia (H

b <1

0 g/

dl)

Surg

ical

inci

sion:

Perip

hera

lU

pper

abd

omin

alIn

trat

hora

cic

Dura

tion

of su

rger

y:

≤2h

>2-3

>3

Emer

genc

y pr

oced

ure

3 16 8 24 17 11 1 15 24 1 16 23 8

Leve

l/Po

int/

Rate

of P

PC

low

/<26

/1.6

%;

med

ium

/26-

44/ 1

3.3%

high

/≥45

/42.

1%(v

alid

ation

subs

ampl

e)

PPC:

Resp

irato

ry in

fecti

on,

resp

irato

ry fa

ilure

, ple

ural

eff

usio

n, a

tele

ctas

is,

pneu

mot

hora

x,

bron

chos

pasm

, asp

iratio

n pn

eum

oniti

s

Deriv

ation

co

hort

AUC:

0.8

9

Valid

ation

co

hort

AUC:

0.8

4

Maz

o et

. al.

2014

2

(Val

idati

on

of A

RISC

AT

scor

e fr

om

Cane

t et.

al.,

2010

1 in

a la

rger

, in

tern

ation

al,

mul

ticen

ter

coho

rt)

Pros

pecti

ve,

mul

ticen

ter,

obse

rvati

onal

co

hort

stud

y

Adul

t pati

ents

un

derg

oing

no

nobs

tetr

ic

surg

ical

pr

oced

ures

un

der g

ener

al,

neur

axia

l, or

pl

exus

blo

ck

anes

thes

ia

5,09

9AR

ISCA

TAs

sess

Re

spira

tory

Ri

sk in

Sur

gica

l Pa

tient

s in

Cata

loni

a

see

abov

eLe

vel/

Poin

t/Pr

edic

ted/

O

bser

ved

Rate

of P

PC

low

/<26

/0.8

7%/ 3

.39%

med

ium

/26-

44/

7.82

%/1

2.96

%hi

gh/≥

45/3

8.13

%/ 3

8.01

%

AUC:

0.8

0

Pred

ictio

n of

sel

ecte

d po

stop

erati

ve p

ulm

onar

y co

mpl

icati

ons

John

son

et.

al. 2

007

14

(Ree

valu

ated

in

a b

road

er

coho

rt fr

om

Aroz

ulla

h et

. al

. 200

0 9 )

Pros

pecti

ve,

mul

ticen

ter,

obse

rvati

onal

co

hort

stud

y

Adul

t pati

ents

un

derg

oing

maj

or

gene

ral o

r vas

cula

r pr

oced

ures

pe

rfor

med

und

er

gene

ral,

spin

al, o

r ep

idur

al a

nest

hesia

90,0

55 d

eriv

ation

89,9

48 v

alid

ation

RRI

Resp

irato

ry

failu

re R

isk

Inde

x

Type

of s

urge

ryIn

tegu

men

tary

Resp

irato

ry a

nd

hem

ic

Hear

tAn

eury

smM

outh

, pal

ate

Stom

ach,

inte

stine

sEn

docr

ine

Pred

ispos

ing

fact

ors

Mal

e se

xAg

e 40

-65

Age

>65

ASA

clas

s 3

ASA

clas

s 4-5

W

ork

RVU

10-

17W

ork

RVU

>17

Em

erge

ncy

Seps

is Hi

stor

y of

seve

re

COPD

As

cite

s Dy

spno

ea

Impa

ired

sens

oriu

m>2

alc

ohol

ic d

rinks

/d

in 2

wk

Blee

ding

diso

rder

sW

eigh

t los

s >10

%Ac

ute

rena

l fai

lure

Cong

estiv

e he

art

failu

re

Smok

er

Stro

ke

Wou

nd c

lass

oth

er

than

cle

an

Preo

pera

tive

albu

min

<3.

5Cr

eatin

ine

>1.5

Pr

eope

rativ

e

1 3 2 2 7 2 2 1 2 2 3 5 2 4 2 2 2 2 2 1 1 1 1 1 2 1 1 1 1 1 1 2

Leve

l/Po

int/

Pred

icte

d/

Obs

erve

d of

Pro

babi

lity

of P

RF

low

/<8/

0.2%

/0.0

8%m

ediu

m/8

-12/

1.0%

/0.8

4%hi

gh/ >

12/6

.6%

/6.7

5%

PRF:

Mec

hani

cal v

entil

ation

fo

r >48

h or

unp

lann

ed

rein

tuba

tion

Deriv

ation

co

hort

AUC:

0.8

56

Valid

ation

co

hort

AUC:

0.8

63

46

Refe

renc

e/Ye

ar

publ

ishe

dSt

udy

desi

gnPa

tient

pop

ulati

onN

umbe

r of

patie

nts

Scor

e ac

rony

mSc

orin

g sy

stem

Cut-

offQ

ualit

y of

pr

edic

tion

bilir

ubin

>1.

0W

hite

blo

od c

ount

<2

.5/ >

10

Preo

pera

tive

seru

m

sodi

um >

145

Plat

elet

cou

nt <

150

SGO

T >4

0 Ha

emat

ocrit

<38

1 1 2 1 1

Brue

ckm

ann

et. a

l. 20

13 15

Retr

ospe

ctive

sin

gle-

cent

re,

obse

rvati

onal

co

hort

stud

y

Case

s with

a

surg

ical

pro

cedu

re

if th

e ad

ult p

atien

t w

as in

tuba

ted

at th

e be

ginn

ing

and

extu

bate

d at

the

end

of th

e pr

oced

ure

33,7

69 o

vera

ll16

,885

der

ivati

on16

,884

val

idati

on

SPO

RCSc

ore

for

Pred

ictio

n of

Po

stop

erati

ve

Resp

irato

ry

Com

plic

ation

s

ASA

scor

e ≥3

Em

erge

ncy

proc

edur

e Hi

gh-r

isk se

rvic

eCo

nges

tive

hear

t fa

ilure

Ch

roni

c pu

lmon

ary

dise

ase

3 3 2 2 1

Scor

e Va

lues

/Pro

babi

lity

of

Rein

tuba

tion

0/0.

12%

1–3/

0.45

%4–

6/1.

64%

7–11

/5.8

6%(v

alid

ation

subs

ampl

e)

Deriv

ation

co

hort

AUC:

0.8

1

Valid

ation

co

hort

AUC:

0.8

1

Pred

ictio

n of

ALI

/ARD

S

Gaj

ic e

t. al

. 20

11 6

(sim

ilar t

o Tr

illo-

Alva

rez

et. a

l. 20

1112

6 , bu

t use

d a

larg

er,

mul

ticen

ter

coho

rt)

Pros

pecti

ve,

mul

ticen

ter,

obse

rvati

onal

co

hort

stud

y

Adul

t pati

ents

w

ith o

ne o

r mor

e AL

I risk

fact

ors,

in

clud

ing

seps

is,

shoc

k, p

ancr

eatiti

s,

pneu

mon

ia,

aspi

ratio

n, h

igh-

risk

trau

ma,

or h

igh-

risk

surg

ery

5,58

4 ov

eral

l2,

500

deriv

ation

3,08

4 va

lidati

on

LIPS

Lung

Inju

ry

Pred

ictio

n Sc

ore

Pred

ispos

ing

Cond

ition

sSh

ock

Aspi

ratio

n Se

psis

Pneu

mon

ia

High

-risk

surg

ery

Ort

hopa

edic

spin

eAc

ute

abdo

men

Card

iac

Aorti

c va

scul

ar

if em

erge

ncy

surg

ery

High

risk

trau

ma

Trau

mati

c br

ain

inju

ry

Smok

e in

hala

tion

Nea

r dro

wni

ng

Lung

con

tusio

nM

ultip

le fr

actu

res

Risk

mod

ifier

sAl

coho

l abu

se

Obe

sity

(BM

I >30

)Hy

poal

bum

inem

iaCh

emot

hera

py

FIO

2>0.

35 (>

4 L/

min

)Ta

chyp

nea

(RR>

30)

SpO

2<95

%

Acid

osis

(pH<

7.35

)Di

abet

es m

ellit

us, i

f se

ptic

2 2 1 1.5

1 2 2.5

3.5

+1.5

2 2 2 1.5

1.5

1 1 1 1 2 1.5

1 1.5

-1

>4 Cut-o

ff fo

r dev

elop

men

t of

ALI/

ARD

S

Com

bine

dAU

C: 0

.80

Sens

itivi

ty:

0.69

Spec

ifici

ty:

0.78

Kor e

t. al

. 20

14 16

(sim

ilar t

o Ko

r et.

al.

2011

127 , b

ut

used

a la

rger

, m

ultic

ente

r co

hort

; se

cond

ary

anal

ysis

of

Gaj

ic e

t. al

. 20

11 6 )

Seco

ndar

y an

alys

is of

apr

ospe

ctive

, m

ultic

ente

r co

hort

stud

y

Adul

t pati

ents

pr

esen

ting

with

on

e or

mor

e AL

I risk

fact

ors,

in

clud

ing

seps

is,

shoc

k, p

ancr

eatiti

s,

pneu

mon

ia,

aspi

ratio

n, h

igh-

risk

trau

ma,

or

high

-risk

surg

ery

and

unde

rgoi

ng a

su

rgic

al p

roce

dure

1,56

2SL

IP 2

Surg

ical

Lu

ng In

jury

Pr

edic

tion

2

Surg

ical

pro

cedu

reHi

gh-r

isk c

ardi

ac

surg

ery

High

-risk

aor

tic

vasc

ular

surg

ery

Emer

genc

y su

rger

yBa

selin

e he

alth

st

atus

Seps

is

Cirr

hosis

Ad

miss

ion

sour

ce

othe

r tha

n ho

me

Phys

iolo

gic

mar

kers

of

acu

te il

lnes

sRe

spira

tory

rate

20

–29

Resp

irato

ry ra

te ≥

30FI

O2 >

35%

Sp

O2 <

95%

7 11 10 10 20 9 7 14 13 5

≥19

Cut-o

ff fo

r dev

elop

men

t of

ARDS

AUC:

0.8

4Se

nsiti

vity

: 0.

82Sp

ecifi

city

: 0.

75

ALI,

acut

e lu

ng in

jury

; ARD

S, a

cute

resp

irato

ry d

istre

ss sy

ndro

me;

ASA

, Am

eric

an S

ocie

ty o

f Ane

sthe

siolo

gist

s cla

ssifi

catio

n; A

UC,

are

a un

der t

he c

urve

; BM

I, bo

dy m

ass i

ndex

; CO

PD,

chro

nic

obst

ructi

ve p

ulm

onar

y di

seas

e; F

IO2,

frac

tion

of in

spire

d ox

ygen

; Hb,

hae

mog

lobi

n; P

PC, p

osto

pera

tive

pulm

onar

y co

mpl

icati

on; P

RF, p

osto

pera

tive

resp

irato

ry fa

ilure

, RVU

, re

lativ

e va

lue

units

(a m

easu

re o

f sur

gica

l com

plex

ity);

SGO

T, se

rum

glu

tam

ic-o

xalo

aceti

c tr

ansa

min

ase;

SpO

2, ox

ygen

satu

ratio

n as

mea

sure

d by

pul

se o

xim

etry

Review intraoperative protective ventilation

Chap

ter

3

47

Refe

renc

e/Ye

ar

publ

ishe

dSt

udy

desi

gnPa

tient

pop

ulati

onN

umbe

r of

patie

nts

Scor

e ac

rony

mSc

orin

g sy

stem

Cut-

offQ

ualit

y of

pr

edic

tion

bilir

ubin

>1.

0W

hite

blo

od c

ount

<2

.5/ >

10

Preo

pera

tive

seru

m

sodi

um >

145

Plat

elet

cou

nt <

150

SGO

T >4

0 Ha

emat

ocrit

<38

1 1 2 1 1

Brue

ckm

ann

et. a

l. 20

13 15

Retr

ospe

ctive

sin

gle-

cent

re,

obse

rvati

onal

co

hort

stud

y

Case

s with

a

surg

ical

pro

cedu

re

if th

e ad

ult p

atien

t w

as in

tuba

ted

at th

e be

ginn

ing

and

extu

bate

d at

the

end

of th

e pr

oced

ure

33,7

69 o

vera

ll16

,885

der

ivati

on16

,884

val

idati

on

SPO

RCSc

ore

for

Pred

ictio

n of

Po

stop

erati

ve

Resp

irato

ry

Com

plic

ation

s

ASA

scor

e ≥3

Em

erge

ncy

proc

edur

e Hi

gh-r

isk se

rvic

eCo

nges

tive

hear

t fa

ilure

Ch

roni

c pu

lmon

ary

dise

ase

3 3 2 2 1

Scor

e Va

lues

/Pro

babi

lity

of

Rein

tuba

tion

0/0.

12%

1–3/

0.45

%4–

6/1.

64%

7–11

/5.8

6%(v

alid

ation

subs

ampl

e)

Deriv

ation

co

hort

AUC:

0.8

1

Valid

ation

co

hort

AUC:

0.8

1

Pred

ictio

n of

ALI

/ARD

S

Gaj

ic e

t. al

. 20

11 6

(sim

ilar t

o Tr

illo-

Alva

rez

et. a

l. 20

1112

6 , bu

t use

d a

larg

er,

mul

ticen

ter

coho

rt)

Pros

pecti

ve,

mul

ticen

ter,

obse

rvati

onal

co

hort

stud

y

Adul

t pati

ents

w

ith o

ne o

r mor

e AL

I risk

fact

ors,

in

clud

ing

seps

is,

shoc

k, p

ancr

eatiti

s,

pneu

mon

ia,

aspi

ratio

n, h

igh-

risk

trau

ma,

or h

igh-

risk

surg

ery

5,58

4 ov

eral

l2,

500

deriv

ation

3,08

4 va

lidati

on

LIPS

Lung

Inju

ry

Pred

ictio

n Sc

ore

Pred

ispos

ing

Cond

ition

sSh

ock

Aspi

ratio

n Se

psis

Pneu

mon

ia

High

-risk

surg

ery

Ort

hopa

edic

spin

eAc

ute

abdo

men

Card

iac

Aorti

c va

scul

ar

if em

erge

ncy

surg

ery

High

risk

trau

ma

Trau

mati

c br

ain

inju

ry

Smok

e in

hala

tion

Nea

r dro

wni

ng

Lung

con

tusio

nM

ultip

le fr

actu

res

Risk

mod

ifier

sAl

coho

l abu

se

Obe

sity

(BM

I >30

)Hy

poal

bum

inem

iaCh

emot

hera

py

FIO

2>0.

35 (>

4 L/

min

)Ta

chyp

nea

(RR>

30)

SpO

2<95

%

Acid

osis

(pH<

7.35

)Di

abet

es m

ellit

us, i

f se

ptic

2 2 1 1.5

1 2 2.5

3.5

+1.5

2 2 2 1.5

1.5

1 1 1 1 2 1.5

1 1.5

-1

>4 Cut-o

ff fo

r dev

elop

men

t of

ALI/

ARD

S

Com

bine

dAU

C: 0

.80

Sens

itivi

ty:

0.69

Spec

ifici

ty:

0.78

Kor e

t. al

. 20

14 16

(sim

ilar t

o Ko

r et.

al.

2011

127 , b

ut

used

a la

rger

, m

ultic

ente

r co

hort

; se

cond

ary

anal

ysis

of

Gaj

ic e

t. al

. 20

11 6 )

Seco

ndar

y an

alys

is of

apr

ospe

ctive

, m

ultic

ente

r co

hort

stud

y

Adul

t pati

ents

pr

esen

ting

with

on

e or

mor

e AL

I risk

fact

ors,

in

clud

ing

seps

is,

shoc

k, p

ancr

eatiti

s,

pneu

mon

ia,

aspi

ratio

n, h

igh-

risk

trau

ma,

or

high

-risk

surg

ery

and

unde

rgoi

ng a

su

rgic

al p

roce

dure

1,56

2SL

IP 2

Surg

ical

Lu

ng In

jury

Pr

edic

tion

2

Surg

ical

pro

cedu

reHi

gh-r

isk c

ardi

ac

surg

ery

High

-risk

aor

tic

vasc

ular

surg

ery

Emer

genc

y su

rger

yBa

selin

e he

alth

st

atus

Seps

is

Cirr

hosis

Ad

miss

ion

sour

ce

othe

r tha

n ho

me

Phys

iolo

gic

mar

kers

of

acu

te il

lnes

sRe

spira

tory

rate

20

–29

Resp

irato

ry ra

te ≥

30FI

O2 >

35%

Sp

O2 <

95%

7 11 10 10 20 9 7 14 13 5

≥19

Cut-o

ff fo

r dev

elop

men

t of

ARDS

AUC:

0.8

4Se

nsiti

vity

: 0.

82Sp

ecifi

city

: 0.

75

ALI,

acut

e lu

ng in

jury

; ARD

S, a

cute

resp

irato

ry d

istre

ss sy

ndro

me;

ASA

, Am

eric

an S

ocie

ty o

f Ane

sthe

siolo

gist

s cla

ssifi

catio

n; A

UC,

are

a un

der t

he c

urve

; BM

I, bo

dy m

ass i

ndex

; CO

PD,

chro

nic

obst

ructi

ve p

ulm

onar

y di

seas

e; F

IO2,

frac

tion

of in

spire

d ox

ygen

; Hb,

hae

mog

lobi

n; P

PC, p

osto

pera

tive

pulm

onar

y co

mpl

icati

on; P

RF, p

osto

pera

tive

resp

irato

ry fa

ilure

, RVU

, re

lativ

e va

lue

units

(a m

easu

re o

f sur

gica

l com

plex

ity);

SGO

T, se

rum

glu

tam

ic-o

xalo

aceti

c tr

ansa

min

ase;

SpO

2, ox

ygen

satu

ratio

n as

mea

sure

d by

pul

se o

xim

etry

48

At the cellular level, physical stimuli are transformed into chemical signals, e.g. pro- and anti-inflammatory mediators by means of direct cell injury or indirect activation of cellular signalling pathways. This process is known as “mechanotransduction”.33 Some mediators may promote local effects such as pro-apoptotic or pro-fibrotic actions, while others act as homing molecules recruiting local and remote immune cell populations (e.g. neutrophils and macrophages).34 These local effects as well as their immunological consequences are summarized by the term “biotrauma”.35

Besides the extracellular matrix, both the endothelial and the epithelial compartment of the alveolar-capillary unit are affected by stress and strain originating from mechanical ventilation. In the endothelium, high stress can lead to direct cell breaks, resulting in capillary stress failure.36,37 Furthermore, mechanical stress as well as inflammatory stimuli (i.e. TNF alpha) may trigger contractions of the cytoskeleton 38 resulting in disruption of adherence junctions,39 which increase endothelial permeability and contribute to oedema formation. Similar to the pulmonary endothelium, mechanical stress and strain increase the permeability of the alveolar epithelium,40 a phenomenon found during ventilation at high,41 as well as low42 lung volumes. Additionally, low lung volume ventilation can lead to repetitive collapse and reopening of lung, affecting the epithelium of small airways, yielding plasma membrane disruption,43 as well as epithelial necrosis and sloughing.44

Figure 1. Alterations of the extracellular matrix in lungs during mechanical ventilation and fluid administration

Review intraoperative protective ventilation

Chap

ter

3

49

Alveolar fluid clearance is essential to maintain intra-alveolar fluid homeostasis, which is usually compromised during VILI. Whereas ventilation with high tidal volumes directly decreases Na,K-adenosintriphosphatase activity,45 ventilation at low lung volumes may indirectly impair fluid clearance due to hypoxia following increased alveolar collapse.46

Impairment of barrier function of the endothelium and epithelium, as well as of fluid clearance leads to the development of interstitial and alveolar oedema, which subsequently causes surfactant dysfunction, and impairs lungs elastic and resistive properties.47 Dysfunction of the surfactant system makes the lung susceptible to alveolar collapse contributing to deterioration of lung mechanics and impairing pulmonary host defense.48

Although most evidence of gross structural alterations of endothelium and epithelium induced by mechanical ventilation originates from in vitro investigations of cultured cells or in vivo investigations in acute lung injury models,49 ventilation applying clinically relevant settings in non-injured lungs can affect the alveolar-capillary barrier function, especially in the presence of independent inflammatory triggers, making mechanical ventilation a powerful hit in presence of systemic inflammation.27

Due to the disturbed integrity of the alveolar-capillary barrier function and consecutive systemic translocation of pathogens or inflammatory mediators, VILI may lead to a systemic inflammatory response affecting not only the lungs, but distal organs as well.50

Lung inhomogeneity, e.g. due to atelectasis formation, is a major contributing factor to the development of VILI. However, most experimental evidence is derived from acute lung injury models. Although their basic pathogenic mechanisms may be similar, the magnitude and time course of atelectasis formation in acute lung injury may be very different from those of atelectasis occurring during anesthesia and relatively short-term intraoperative mechanical ventilation. Resorption of alveolar gas51,52 and compression of lung structures53-56 may lead to atelectasis during short-term mechanical ventilation in non-injured lungs, whereby the former might play a more important role.

In a porcine model of experimental pneumonia, both exogenous surfactant administration and ventilation according to the open lung approach attenuated bacterial growth and systemic translocation by minimizing alveolar collapse and atelectasis formation.57 In a similar model of experimental pneumonia in mechanically ventilated piglets, bacterial translocation was lowest with individually tailored PEEP levels, whereas low and high PEEP promoted bacterial translocation.58

In isolated non-perfused mouse lungs, both an “open lung approach” (tidal volume 6 mL/kg, recruitment manoeuvres and PEEP of 14 - 16 cmH2O) as well as a “lung rest strategy” (tidal volume of 6 ml/kg, PEEP of 8 - 10 cmH2O, no recruitment manoeuvres) were associated with reduced pulmonary inflammatory response and improved respiratory mechanics compared to injurious mechanical ventilation (tidal volume of 20 ml/kg, PEEP of 0 cmH2O).59 Interestingly, the “lung rest strategy” was associated with less apoptosis but more ultrastructural cell damage, most likely due to increased activation of mitogen-activated protein kinase pathways as compared to the “open lung strategy”.59

50

In healthy mice, mechanical ventilation with a tidal volume of 8 ml/kg and PEEP of 4 cmH2O induced a reversible increase in plasma and lung tissue cytokines as well as increased leukocyte influx, but the integrity of the lung tissue was preserved.60 In another investigation, even least-injurious ventilator settings were able to induce VILI in the absence of a previous pulmonary insult in mice.61 Of note, the deleterious effects of mechanical ventilation in non-injured lungs are partly dependent on its duration.62 However, an experimental study demonstrated that large tidal volumes had only minor if any deleterious effects on lungs, despite prolonged mechanical ventilation.23 Possibly, this is explained by the lack of a previous inflammatory insult, as for example surgery. In fact, systemic inflammation may prime the lungs to injury by mechanical ventilation.63

Mechanical ventilation strategies to protect lungs during surgeryAtelectasis and intraoperative mechanical ventilationAtelectasis develops in as much as 90% of patients undergoing general anesthesia,64 and can persist to different degrees after surgery, also surrounding pleura effusion, as illustrated in figure 2. The area of non-aerated lung tissue near to the diaphragm varies depending on the surgical procedure and patient characteristics, but has been estimated in the range of 3-6 %65-67 to 20-25%,64 and even higher if calculated as amount of tissue.

Different mechanisms have been postulated to favour atelectasis formation during anesthesia, including: 1) collapse of small airways;68-70 2) compression of lung structures;53-56 3) absorption

Figure 2. Magnetic resonance imaging (MRI) scans of lungs of three patients before and on the first day after open abdominal surgery

Review intraoperative protective ventilation

Chap

ter

3

51

of intra-alveolar gas content;51,52 and 4) impairment of lung surfactant function.71 Mechanical ventilation strategies for general anesthesia have been importantly influenced by the progressive decrease in oxygenation and compliance.72 Tidal volumes up to 15 mL/kg of predicted body weight were advocated to increase the end-expiratory lung volume (EELV) and counteract atelectasis in the intraoperative period.72 Provided there is no contraindication, PEEP and lung recruitment manoeuvres may also contribute to revert or prevent the loss of EELV and closure of small airways during anesthesia.

Tidal volumes for intraoperative protective ventilationDriven by clinical and experimental studies, tidal volumes during mechanical ventilation have been importantly reduced in patients suffering from the acute respiratory distress syndrome (ARDS) in order to limit lung overdistension.73 Influenced by this practice in intensive care unit patients, a similar trend was observed in the operation room. As reported by different investigators,74,75 average tidal volumes in the range of 6 to 9 mL/kg of predicted body weight have gained broad acceptance for non-injured lungs, in spite of experimental23,76 and clinical data77-

79 suggest that higher values are not associated with increased lung damage or inflammation. Furthermore, anaesthesiologists have consistently reduced tidal volumes also during one-lung ventilation. Whereas values as high as 10 mL/kg have been used in the past, experimental80,81 and clinical82-85 studies have suggested that tidal volumes of approximately 4 to 5 mL/kg may be more appropriate for lung protection, while still allowing adequate gas exchange. Furthermore, a small RCT showed that atelectasis did not increase significantly with low tidal volume without PEEP from induction of anesthesia until the end of surgery.65 This is also supported by the fact that mechanical ventilation with low tidal volume and PEEP did not result in a progressive deterioration of the respiratory system compliance and gas exchange during open abdominal surgery in a larger RCT.86 It must be kept in mind that “set” and “actual” (i.e. delivered) tidal volumes can differ substantially,87 and that settings should be adjusted judiciously.

Positive end-expiratory pressure for intraoperative protective ventilationClinical studies have shown that a PEEP of 10 cmH2O is required to reduce or eliminate atelectasis,67,88,89 improve compliance without increasing deadspace,90,91 and maintain end-expiratory lung volume during general anesthesia in both non-obese and obese patients.92 Another study in normal subjects showed that PEEP of 10 cmH2O increased lung volume, but did not improve the respiratory function compared to PEEP of 0 cmH2O.54 Certainly, the level of PEEP should be chosen according to the patient’s particular characteristics, the particularities of the surgical approach, and patient positioning. Several targets have been proposed for a more individual titration of PEEP during general anesthesia, including the following: 1) oxygenation,93 also combined with dead space,90 or EELV91 ; 2) mechanical properties of the respiratory system94 ; and 3) distribution of ventilation using electric impedance tomography.95,96 However, none of these has been shown to improve patient outcome.

Although controversial, an alternative approach for PEEP during general anesthesia is the so-called “intraoperative permissive atelectasis”, when PEEP is kept relatively low and recruitment manoeuvres are waived. This concept aims at reducing the static stress in lungs, which is closely related to the mean airway pressure, assuming that collapsed lung tissue is protected against injury from mechanical ventilation (figure 3). Intraoperative permissive atelectasis may be limited

52

Figure 3. Effects of high and low tidal volumes (VT) at end-inspiration and end-expiration with low and high PEEP during general anesthesia

by deterioration in oxygenation, which could require higher inspiratory oxygen fractions. Also, shear stress may occur at the interface between collapsed and open tissue,18 likely resulting in lung damage and inflammation, even in presence of low global stress.97 Theoretically, intraoperative low PEEP could increase the incidence and the amount of atelectasis even in the postoperative period, resulting in further PPCs. A recent large retrospective study investigating the association between intraoperative mechanical ventilator settings and outcomes suggested that the use of “minimal” PEEP (2.2 to 5 cmH2O) combined with low tidal volumes (6 to 8 mL/kg) is associated with increased risk of 30-day mortality.77 However, a large international multicenter RCT challenged the concept that “minimal” PEEP combined with low tidal volumes in the intraoperative period is harmful.86 Also in elderly patients undergoing major open abdominal surgery, a strategy consisting of low tidal volume, PEEP 12 cmH2O and recruitment manoeuvres increased the PaO2 intraoperatively compared to a strategy with high tidal volume without PEEP, but this effect was not maintained in the postoperative period.98 Even without recruitment manoeuvres PEEP improved oxygenation during upper abdominal surgery compared to zero end-expiratory pressure, but again such effects were limited to the intraoperative period and did not prevent postoperative complications.99

Lung recruitment manoeuvres for intraoperative protective ventilationPEEP is most effective for preserving respiratory function if preceded by a recruitment manoeuvre, which must overcome the opening pressures of up to 40 cmH2O in non-obese,100 and 40-50 cmH2O in obese patients,101 in the absence of lung injury. Recruitment manoeuvres can be performed in different ways using the anesthesia ventilator, as illustrated in figure 4.

Review intraoperative protective ventilation

Chap

ter

3

53

Most commonly, such manoeuvres are performed by “bag squeezing” using the airway pressure-limiting valve of the anesthesia machine (figure 4A). However, recruitment manoeuvres are better controlled if performed during tidal ventilation, for example using a stepwise increase of PEEP, tidal volumes, or a combination of these (figure 4B). Provided there are no contraindications, the inspiratory plateau pressure as high as 40 cmH2O, are more likely to result in full recruitment.102

In anesthesia devices that allow pressure-controlled ventilation, recruitment manoeuvres can be conducted with a constant driving pressure of 15-20 cmH2O, and by increasing PEEP up to 20 cmH2O in steps of 5 cmH2O (30 to 60 s per step). After three to five breaths at a PEEP level that allows achieving the target inspiratory pressure, PEEP and tidal volume are adjusted to the respective desired levels (figure 4C).

A

B

C

Figure 4. Illustrative fluctuation of airway pressure during three types of lung recruitment manoeuvres for intraoperative mechanical ventilation

54

Tabl

e 3.

Ran

dom

ized

con

trol

led

tria

ls u

sing

non

clin

ical

prim

ary

outc

omes

Refe

renc

e/Ye

ar

publ

ishe

dSt

udy

desi

gnPa

tient

po

pula

tion/

N

umbe

rIn

terv

entio

n gr

oup(

s)Co

ntro

l gro

upN

oncl

inic

al p

rimar

y ou

tcom

esSe

cond

ary

outc

omes

Thor

acic

sur

gery

Wrig

ge e

t al.

2004

78

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

m

ajor

thor

acic

su

rger

yn=

34 (2

ex

clud

ed)

PV V T: 6

mL/

kgPE

EP: 1

0 cm

H 2O

P aw

Lim

it 35

cm

H 2O

durin

g TL

V an

d O

LVn=

15

CV V T: 1

2-15

mL/

kgZE

EPP a

w L

imit

35 c

mH 2

Odu

ring

TLV

and

OLV

n=17

Infla

mm

ator

y m

edia

tors

in

pla

sma:

no

diffe

renc

es

betw

een

grou

ps fo

r TN

Fα, I

L-1,

IL

-6, I

L-8,

IL-1

0, IL

-12

Gas

exc

hang

e: n

o di

ffere

nces

bet

wee

n gr

oups

Schi

lling

et a

l. 20

05 82

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

el

ectiv

e op

en

thor

acic

surg

ery

n=32

PV V T: 5

mL/

kgdu

ring

TLV

and

OLV

PEEP

: 3 c

mH 2

O, T

LVPE

EP: 0

-2 c

mH 2

O, O

LVP a

w L

imit

30 c

mH 2

On=

16

CV V T: 1

0 m

L/kg

durin

g TL

V an

d O

LVPE

EP: 3

cm

H 2O

, TLV

PEEP

: 0-2

cm

H 2O

, OLV

P aw

Lim

it 30

cm

H 2O

n=16

Infla

mm

ator

y m

edia

tors

in

BAL:

TN

Fα a

nd sI

CAM

low

er d

urin

g PV N

o di

ffere

nces

bet

wee

n gr

oups

fo

r cel

l cou

nt, P

MN

ela

stas

e,

tota

l pro

tein

, alb

umin

, IL-

8,

IL-1

0

PaO

2/FI

O2:

no

diffe

renc

es b

etw

een

grou

psPa

CO2:

high

er d

urin

g PV

Mic

hele

t et a

l. 20

06 83

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

pl

anne

d es

opha

gect

omy

n=52

PV V T: 9

mL/

kg, T

LVV T

: 5 m

L/kg

OLV

PEEP

: 5 c

mH 2

O, T

LV

and

OLV

n=26

CV V T: 9

mL/

kg, T

LV a

nd O

LVZE

EP, T

LV a

nd O

LV

n=26

Infla

mm

ator

y m

edia

tors

in

plas

ma:

IL-1

ß, IL

-6, I

L-8

low

er

durin

g PV

No

diffe

renc

es b

etw

een

grou

ps

for T

NFα

PaO

2/FI

O2

and

PaCO

2: hi

gher

dur

ing

PVEV

LWI:

low

er d

urin

g PV

Tim

e to

ext

ubati

on:

shor

ter d

urin

g PV

Lin

et a

l. 20

08 10

3

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

es

opha

gect

omy

n=40

PV V T: 1

0 m

L/kg

, TLV

V T: 5

-6 m

L/kg

OLV

PEEP

: 3-5

cm

H 2O

, OLV

n=20

CV V T: 1

0 m

L/kg

, TLV

and

OLV

ZEEP

, TLV

and

OLV

n=20

Infla

mm

ator

y m

edia

tors

in

plas

ma:

IL-6

, IL-

8, lo

wer

dur

ing

PV

P pea

k, P p

lat,

and

R aw

: lo

wer

dur

ing

PV

Review intraoperative protective ventilation

Chap

ter

3

55

Unz

ueta

et a

l. 20

12 10

4

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

el

ectiv

e op

en

thor

acot

omy

n=40

PV V T: 8

mL/

kg, T

LVV T

: 6 m

L/kg

OLV

PEEP

: 8 c

mH 2

O, T

LV

and

OLV

RM w

ith st

epw

ise

PEEP

/ Paw

incr

ease

un

til 2

0/40

cm

H 2O

be

fore

star

t of O

LVn=

20

CV V T: 8

mL/

kg, T

LVV T

: 6 m

L/kg

OLV

PEEP

: 8 c

mH 2

O, T

LV a

nd

OLV

no R

M b

efor

e st

art o

f OLV

n=20

Dead

spac

e: lo

wer

dur

ing

PVPa

O2/

FIO

2: hi

gher

du

ring

PVPa

CO2:

low

er d

urin

g PV

Card

iac

surg

ery

Kone

r et a

l. 20

04 10

5

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

el

ectiv

e on

-pum

p co

rona

ry a

rter

y by

pass

gra

fting

su

rger

yn=

44

(1) P

VV T

: 6 m

L/kg

PEEP

: 5 c

mH 2

On=

15(2

) CV+

PEEP

V T: 1

0 m

L/kg

PEEP

: 5 c

mH 2

On=

14

(3) C

V+ZE

EPV T

: 10

mL/

kgPE

EP: 0

cm

H 2O

n=15

Infla

mm

ator

y m

edia

tors

in

pla

sma:

no

diffe

renc

es

betw

een

grou

ps fo

r TN

Fα a

nd

IL-6

P pla

t: lo

wer

dur

ing

PV

com

pare

d to

bot

h CV

gr

oups

Shun

t fra

ction

: low

er

durin

g PV

com

pare

d to

bo

th C

V+ZE

EPPa

O2/

FIO

2: hi

gher

du

ring

venti

latio

n w

ith

PEEP

, (1)

+ (2

)

Zupa

ncic

h et

al.

2005

106

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

el

ectiv

e on

-pum

p co

rona

ry a

rter

y by

pass

gra

fting

su

rger

yn=

40

PV V T: 8

mL/

kgPE

EP: 1

0 cm

H 2O

n=20

CV V T: 1

0-12

mL/

kgPE

EP: 2

-3 c

mH 2

On=

20

Infla

mm

ator

y m

edia

tors

in

plas

ma

and

BAL:

IL-6

, IL-

8,

low

er d

urin

g PV

in b

oth.

PaCO

2: hi

gher

dur

ing

PV

Reis

Mira

nda

et

al. 2

005

93

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

el

ectiv

e on

-pum

p co

rona

ry a

rter

y by

pass

gra

fting

or

val

ve su

rger

yn=

62

(1) L

ate

open

lung

V T: 4

-6 m

L/kg

PEEP

: 10

cmH 2

Ost

artin

g at

pos

top

ICU

ar

rival

n=18

(2) E

arly

ope

n lu

ngV T

: 4-6

mL/

kgPE

EP: 1

0 cm

H 2O

star

ting

after

in

tuba

tion

n=22

(3) V

T: 6-

8 m

L/kg

PEEP

: 5 c

mH 2

On=

22

Infla

mm

ator

y m

edia

tors

in

plas

ma:

IL-8

dec

reas

ed a

fter

CPB

in b

oth

open

lung

gro

ups;

IL

-10

decr

ease

d fa

ster

afte

r CP

B in

ear

ly o

pen

lung

gro

up

Evid

ence

of

perio

pera

tive

myo

card

ial i

nfar

ction

(c

k-M

B an

d EC

G):

no

diffe

renc

es b

etw

een

grou

ps

56

Abdo

min

al s

urge

ry

Wrig

ge e

t al.

2004

78

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

m

ajor

abd

omin

al

surg

ery

n=30

PV V T: 6

mL/

kgPE

EP: 1

0 cm

H 2O

P aw

Lim

it 35

cm

H 2O

n=15

CV V T: 1

2-15

mL/

kgZE

EPP a

w L

imit

35 c

mH 2

On=

15

Infla

mm

ator

y m

edia

tors

in

pla

sma:

no

diffe

renc

es

betw

een

grou

ps fo

r TN

Fα, I

L-1,

IL

-6, I

L-8,

IL-1

0, IL

-12

PaO

2/FI

O2:

no

diffe

renc

es b

etw

een

grou

psPa

CO2:

high

er d

urin

g PV

Wol

thui

s et a

l. 20

08 10

7

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

a

surg

ical

pr

oced

ure

in g

ener

al

anes

thes

ia ≥

5hn=

46

PV V T: 6

mL/

kgPE

EP: 1

0 cm

H 2O

n=24

CV V T: 1

0-12

mL/

kgZE

EPn=

22

Infla

mm

ator

y m

edia

tors

in

pla

sma

and

BAL:

low

er

mye

lope

roxi

dase

and

nu

cleo

som

e le

vel i

n BA

L du

ring

PV

PaCO

2: hi

gher

dur

ing

PVPP

Cs: n

o di

ffere

nces

be

twee

n gr

oups

Wei

ngar

ten

et

al. 2

010

98

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

ag

ed >

65y

unde

rgoi

ng

maj

or o

pen

abdo

min

al

surg

ery

unde

r ge

nera

l an

esth

esia

n=40

PV V T: 6

mL/

kgPE

EP: 1

2 cm

H 2O

RM w

ith st

epw

ise

PEEP

incr

ease

unti

l 15

cmH 2

On=

20

CV V T: 1

0 m

L/kg

ZEEP

no R

Mn=

20

Infla

mm

ator

y m

edia

tors

in

pla

sma:

no

diffe

renc

es

betw

een

grou

ps

PaO

2/FI

O2 +

PaC

O2:

high

er d

urin

g PV

Com

plia

nce

high

er a

nd

resis

tanc

e lo

wer

dur

ing

PV

Spin

al s

urge

ry

Mem

tsou

dis

et

al. 2

012

108

Pros

pecti

ve,

singl

e-ce

ntre

, ra

ndom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

el

ectiv

e lu

mba

r de

com

pres

sion

and

fusio

n in

pr

one

positi

on

unde

r gen

eral

an

esth

esia

n=26

PV V T: 6

mL/

kgPE

EP: 8

cm

H 2O

n=13

CV V T: 1

2 m

L/kg

ZEEP

n=13

Infla

mm

ator

y m

edia

tors

in

pla

sma:

no

diffe

renc

es

betw

een

grou

psPa

CO2:

high

er d

urin

g PV

BAL,

bro

ncho

alve

olar

lava

ge; C

K-M

B, m

uscl

e-br

ain

type

cre

atine

kin

ase;

CPB

, car

diop

ulm

onar

y by

pass

; CV,

con

venti

onal

ven

tilati

on; E

CG, e

lect

roca

rdio

gram

; EVL

WI,

extr

avas

cula

r lun

g w

ater

inde

x; F

IO2,

insp

ired

fracti

on o

f oxy

gen;

ICAM

, int

erce

llula

r adh

esio

n m

olec

ule;

ICU,

inte

nsiv

e ca

re u

nit;

IL, i

nter

leuk

in; P

aw, a

irway

pre

ssur

e; P

peak

. pea

k pr

essu

re; P

plat

, pla

teau

pre

ssur

e;

PaCO

2, pa

rtial

pre

ssur

e of

art

eria

l car

bon

diox

ide;

PaO

2, pa

rtial

pre

ssur

e of

art

eria

l oxy

gen;

PaO

2/FI

O2,

ratio

of p

artia

l pre

ssur

e of

art

eria

l oxy

gen

to in

spire

d fra

ction

of o

xyge

n; P

EEP,

positi

ve

end–

expi

rato

ry p

ress

ure;

PM

N, p

olym

orph

onuc

lear

leuk

ocyt

e; P

PCs,

post

oper

ative

pul

mon

ary

com

plica

tions

; PV,

pro

tecti

ve ve

ntila

tion;

OLV

, one

-lung

venti

latio

n; R

aw, a

irway

resis

tanc

e; R

M,

recr

uitm

ent m

anoe

uvre

; sIC

AM, s

olub

le in

terc

ellu

lar a

dhes

ion

mol

ecul

e; T

NFα,

tum

our n

ecro

sis fa

ctor

alp

ha; T

LV, t

wo-

lung

ven

tilati

on; V

T, tid

al v

olum

e; Z

EEP,

zero

end

–exp

irato

ry p

ress

ure

Review intraoperative protective ventilation

Chap

ter

3

57

Recent evidence for intraoperative protective ventilation Randomized controlled trials using non-clinical primary outcomesThe literature search identified eleven RCTs that compared a protective ventilation strategy with a non-protective ventilation strategy during general anesthesia for surgery with regard to non-clinical primary outcome in patients undergoing thoracic surgery,78,82,83,103,104 cardiac surgery,93,105,106 abdominal surgery,78,98,107 or spinal surgery,108 as depicted in table 3. In eight RCTs the protective ventilation strategy consisted of both lower tidal volumes and higher levels of PEEP;78,83,98,103,105-108 in two RCTs, it consisted of either lower tidal volume,82 a higher level of PEEP.93 In one RCT, lung recruitment manoeuvres were used during the protective ventilation strategy.104

The effects on inflammatory responses are slightly contradictory. While four RCTs showed no difference in local levels of inflammatory mediators between patients on protective and those on non-protective ventilation,78,98,105,108 six RCTs showed that protective strategies were associated with lower levels of inflammatory mediators.82,83,93,103,106,107

Randomized controlled trials using clinical primary outcomesIn total, 8 RCTs were identified that compared a protective ventilation strategy with a non-protective ventilation strategy during surgery with regard to a clinical primary outcomes in patients planned for abdominal surgery,86,109-111 thoracic surgery,85,112 cardiac surgery,113 or spinal surgery,114 as shown in table 4. In four RCTs the protective strategy consisted of both lower tidal volumes and higher levels of PEEP,110-112,114 in the four remaining RCTs it consisted of either lower tidal volumes,85,109,113 or higher levels of PEEP.86

Four trials reported on PPCs in the first postoperative days, including bronchitis, hypoxemia, and atelectasis,114 pneumonia, need for invasive or non-invasive ventilation for acute respiratory failure,110 a modified “Clinical Pulmonary Infection Score” and chest x–ray abnormalities,111 and hypoxemia, bronchospasm, suspected pulmonary infection, pulmonary infiltrate, aspiration pneumonitis, development of ARDS, atelectasis, pleural effusion, pulmonary oedema, and pneumothorax.86

In a Chinese single-centre RCT,114 investigators compared protective ventilation (tidal volume 6 mL/kg and 10 cmH2O PEEP) versus non-protective (tidal volume 10 to 12 mL/kg and 0 cmH2O PEEP) in 60 elderly American Society of Anesthesiologists class II and III patients scheduled for spinal surgery. Patients receiving protective ventilation had less PPCs.

In a French multi-centre trial (Intraoperative PROtective VEntilation, IMPROVE),110 protective ventilation (tidal volume 6 to 8 mL/kg and PEEP 6 to 8 cmH2O) was compared with non-protective ventilation (tidal volume 10 to 12 mL/kg and 0 cmH2O PEEP in 400 non-obese patients at intermediate to high risk of pulmonary complications after planned major abdominal surgery. The primary outcome (postoperative pulmonary and extra–pulmonary complications) occurred less often in patients receiving ‘protective’ ventilation. Such complications have been ascribed to the release of inflammatory mediators by the lungs into the systemic circulation, affecting the lungs,115 as well as peripheral organs.50 These patients also had a shorter length of hospital stay, but mortality was unaffected.

58

Tabl

e 4.

Ran

dom

ized

con

trol

led

tria

ls u

sing

clin

ical

prim

ary

outc

omes

Refe

renc

e/Ye

ar p

ublis

hed

Stud

y de

sign

Patie

nt p

opul

ation

/ N

umbe

rIn

terv

entio

nCo

ntro

l gro

upCl

inic

al p

rimar

y ou

tcom

esSe

cond

ary

outc

omes

Thor

acic

sur

gery

Mas

low

et.

al.

2013

112

Pros

pecti

ve, s

ingl

e-ce

ntre

, ran

dom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

ele

ctive

pulm

onar

y re

secti

on=3

4

PV V T: 5

mL/

kg, T

LV a

nd

OLV

PEEP

: 5 c

mH 2

O, T

LV

and

OLV

n=17

CV V T: 1

0 m

L/kg

, TLV

an

d O

LVZE

EP, T

LV a

nd O

LVn=

17

Rate

of a

tele

ctas

is:

low

er w

ith C

V;le

ngth

of h

ospi

tal

stay

: no

diffe

renc

es

betw

een

grou

ps

PaCO

2 abd

dea

d sp

ace:

hig

her

durin

g PV

C dyn

: hig

her d

urin

g CV

Shen

et a

l. 20

13 85

Pros

pecti

ve, s

ingl

e-ce

ntre

, ran

dom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

ele

ctive

th

orac

osco

pic

esop

hage

ctom

yn=

101

PV V T: 8

mL/

kg, T

LVV T

: 5 m

L/kg

OLV

PEEP

: 5 c

mH 2

O, T

LV

and

OLV

n=53

CV V T: 8

mL/

kg, T

LVV T

: 8 m

L/kg

OLV

ZEEP

, TLV

and

OLV

n=48

PPCs

: low

er ra

te

with

PV

Mor

talit

y: n

o di

ffere

nce

betw

een

grou

ps

PaO

2/FI

O2

and

PaCO

2: hi

gher

du

ring

PVIn

flam

mat

ory

med

iato

rs in

BA

L: lo

wer

IL-1

ß, IL

-6 a

nd IL

-8

Card

iac

surg

ery

Sund

ar e

t al.

2011

11

3

Pros

pecti

ve, s

ingl

e-ce

ntre

, ran

dom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

ele

ctive

ca

rdia

c su

rger

yn=

149

PV V T: 6

mL/

kgPE

EP/F

IO2:

acco

rdin

g to

ARD

S N

etw

ork

tabl

en=

75

CV V T: 1

0 m

L/kg

PEEP

/FIO

2: ac

cord

ing

to

ARDS

Net

wor

k ta

ble

n=74

Rate

of r

eint

ubati

on:

low

er w

ith P

VN

umbe

r of p

atien

ts

requ

iring

ven

tilati

on

6h p

osto

pera

tivel

y:

low

er w

ith P

V

Gas

exc

hang

e: n

o di

ffere

nce

betw

een

grou

ps

Abdo

min

al s

urge

ry

Tres

chan

et a

l. 20

12 10

9

Pros

pecti

ve, s

ingl

e-ce

ntre

, ran

dom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

ele

ctive

up

per a

bdom

inal

surg

ery

≥3 h

und

er c

ombi

ned

gene

ral a

nd e

pidu

ral

anes

thes

ian=

101

PV V T: 6

mL/

kgPE

EP: 5

cm

H 2O

n=50

CV V T: 1

2 m

L/kg

PEEP

: 5 c

mH 2

On=

51

Rate

of a

tele

ctas

is:

low

er w

ith C

V

PaO

2/FI

O2:

high

er d

urin

g CV

C dyn

and

Raw

: hig

her d

urin

g CV

PaO

2 at p

osto

pera

tive

day

5:

high

er w

ith C

V

Review intraoperative protective ventilation

Chap

ter

3

59

Futie

r et a

l. 20

13

110

Pros

pecti

ve

rand

omize

d co

ntro

lled

mul

ti-ce

ntre

stud

y

Adul

ts p

atien

ts a

t in

term

edia

te to

hig

h ris

k of

pul

mon

ary

com

plic

ation

s un

derg

oing

maj

or

abdo

min

alsu

rger

yn=

400

PV V T: 6

-8 m

L/kg

PEEP

: 6-8

cm

H 2O

n=20

0

CV V T: 1

0-12

mL/

kgZE

EPn=

200

Com

posit

e pr

imar

y ou

tcom

e of

maj

or

pulm

onar

y or

ex

trap

ulm

onar

y co

mpl

icati

ons:

low

er

with

PV

Redu

ced

rate

of a

tele

ctas

is,

pneu

mon

ia, n

eed

for

venti

latio

n w

ithin

7 d

ays a

nd

seps

is w

ith P

V.Re

duce

d le

ngth

of h

ospi

tal

stay

with

PV

Seve

rgni

ni e

t al.

2013

111

Pros

pecti

ve, s

ingl

e-ce

ntre

, ran

dom

ized

cont

rolle

d tr

ial

Adul

t pati

ents

un

derg

oing

ele

ctive

op

en a

bdom

inal

surg

ery

≥2 h

n=56

(1 e

xclu

ded)

PV V T: 7

mL/

kgPE

EP: 1

0 cm

H 2O

n=28

CV V T: 9

mL/

kgZE

EPn=

27

Pulm

onar

y fu

nctio

n te

sts:

impr

oved

with

PV

Mod

ified

Clin

ical

Pul

mon

ary

Infe

ction

Sco

re: l

ower

with

PV

PaO

2 at

pos

tope

rativ

e da

ys 1

, 3,

5: h

ighe

r with

PV

Rate

of c

hest

x-r

ay

abno

rmal

ities

: low

er w

ith P

VLe

ngth

of h

ospi

tal s

tay:

no

diffe

renc

e be

twee

n gr

oups

PRO

VE N

etw

ork

Inve

stiga

tors

20

14 86

Pros

pecti

ve,

inte

rnati

onal

, m

ultic

ente

r, ra

ndom

ized

cont

rolle

d tr

ial

Adul

ts p

atien

ts a

t in

term

edia

te to

hig

h ris

k of

pul

mon

ary

com

plic

ation

s un

derg

oing

maj

or

abdo

min

alsu

rger

yn=

900

PV V T: 8

mL/

kgPE

EP: 1

2 cm

H 2O

RM w

ith st

epw

ise

incr

ease

of V

T afte

r in

ducti

on a

nd b

efor

e ex

tuba

tion

n=44

5

CV V T: 8

mL/

kgPE

EP: 0

-2 c

mH 2

On=

449

Rate

of P

PCs:

no

diffe

renc

e be

twee

n gr

oups

Rate

of i

ntra

oper

ative

hy

pote

nsio

n an

d am

ount

of

vaso

activ

e dr

ugs g

iven

: hig

her

durin

g PV

Rate

of d

esat

urati

on: l

ower

du

ring

PVM

orta

lity

and

leng

th o

f ho

spita

l sta

y: n

o di

ffere

nce

betw

een

grou

ps

Spin

al s

urge

ry

Ge

et a

l. 20

13 11

4Pr

ospe

ctive

, sin

gle-

cent

re, r

ando

mize

d co

ntro

lled

tria

l

Adul

t pati

ents

un

derg

oing

spin

e fu

sion

n=60

PV V T: 6

mL/

kgPE

EP: 1

0 cm

H 2O

RM e

very

15m

inn=

30

CV V T: 1

0-12

mL/

kgZE

EPn=

30

Rate

of P

PCs,

low

er

with

PV

PaO

2/FI

O2,

high

er d

urin

g PV

ARDS

, acu

te re

spira

tory

dist

ress

synd

rom

e; B

AL, b

ronc

hoal

veol

ar la

vage

; CV,

con

venti

onal

ven

tilati

on; F

IO2,

insp

ired

frac

tion

of o

xyge

n; IC

U, in

tens

ive

care

uni

t; IL

, int

erle

ukin

; PaC

O2,

parti

al p

ress

ure

of a

rter

ial c

arbo

n di

oxid

e; P

aO2,

parti

al p

ress

ure

of a

rter

ial o

xyge

n; P

aO2/

FIO

2, ra

tio o

f par

tial p

ress

ure

of a

rter

ial o

xyge

n to

insp

ired

frac

tion

of o

xyge

n; P

EEP,

pos

itive

en

d–ex

pira

tory

pre

ssur

e; P

PCs,

pos

tope

rativ

e pu

lmon

ary

com

plic

ation

s; P

V, p

rote

ctive

ven

tilati

on; O

LV, o

ne-lu

ng v

entil

ation

; Raw

, airw

ay re

sista

nce;

RM

, rec

ruitm

ent m

anoe

uvre

; TLV

, tw

o-lu

ng v

entil

ation

; VT,

tidal

vol

ume;

ZEEP

, zer

o en

d–ex

pira

tory

pre

ssur

e

60

Figu

re 5

. Odd

s ra

tios

for p

osto

pera

tive

pulm

onar

y co

mpl

icati

ons

of “

prot

ectiv

e” v

ersu

s “n

on-p

rote

ctive

” ve

ntila

tion

in tr

ials

Review intraoperative protective ventilation

Chap

ter

3

61

An Italian single–centre trial111 investigated the effectiveness of protective ventilation (tidal volume 7 mL/kg and 10 cmH2O PEEP with recruitment manoeuvres) versus non–protective ventilation (tidal volume 9 mL/kg and zero end-expiratory pressure) in 56 patients scheduled for open abdominal surgery lasting more than 2 hours. The modified “Clinical Pulmonary Infection Score” was lower in patients receiving protective ventilation. These patients also had fewer chest x–ray abnormalities and higher arterial oxygenation compared to patients receiving non-protective ventilation.

Finally, in an international multicenter trial conducted in Europe and the Americas (PROtective Ventilation using HIgh versus LOw PEEP, PROVHILO),86 the PROtective VEntilation (PROVE) Network–investigators compared PEEP of 12 cmH2O combined with recruitment manoeuvres versus PEEP of 2 cmH2O without recruitment manoeuvres in 900 non-obese patients at high risk for postoperative pulmonary complications planned for open abdominal surgery under ventilation at tidal volumes of 8 mL/kg. The incidence of PPCs was not different between patients receiving protective ventilation and patients receiving non-protective ventilation.

Challenges of studies using bundles As shown in preceding subsections “Randomized controlled trials using non-clinical primary outcomes” and “Randomized controlled trials using clinical primary outcomes”, most RCTs addressing intraoperative mechanical ventilation compared bundles of interventions consisting of tidal volumes and levels of PEEP, usually accompanied by a lung recruitment maneuver.110-112,114 Notably, recruitment manoeuvres differed between the trials. In the Italian single-centre RCT,111 investigators used incremental titration of tidal volumes until a plateau pressure of 30 cmH2O, directly after induction of anesthesia, after any disconnection from the ventilator and immediately before extubation, similar as in PROVHILO.86 In IMPROVE,110 recruitment was performed with a continuous positive airway pressure of 30 cmH2O for 30 seconds every 30 minutes, also known as sustained inflation, after tracheal intubation. Finally, in the Chinese single-centre RCT,114 the recruitment manoeuvres followed a similar approach, but to plateau pressures of up to 35 cmH2O, and they were performed every 15 minutes. It is difficult, if not impossible to conclude from these trials what caused the benefit: tidal volume reduction or increase of PEEP, or both, and to determine the role of recruitment manoeuvres. Moreover, to what extent the recruitment manoeuvre has succeeded in reopening lung has not been analysed in the different studies.

The results of the PROVHILO trial, however, suggest that low tidal volumes rather than PEEP combined with lung recruitment manoeuvres are responsible for lung protection in the intraoperative period. This interpretation is also supported by an analysis of different studies on the odds ratios of lower tidal volumes,85,109,113 higher levels of PEEP,86 as well as their combination,110-112,114 regarding the development of PPCs (figure 5).

Certainly, these conclusions are only valid for the studied population, that is, non-obese patients at risk of PPCs undergoing elective abdominal surgery. Other patient populations could still benefit from higher levels of PEEP and recruitment manoeuvres.

Challenges of composite outcome measuresComposite outcome measures offer the benefit of an increased event rate, which is helpful

62

to ensure adequate statistical power of a trial.8 It is reasonable to combine related outcomes that represent different aspects of a single underlying pathophysiological process, like PPCs for VILI. There are, though, two major limitations regarding the use of composite outcomes. First, the component variables can differ importantly in terms of severity and frequency. Second, differences in the frequency of component variables in a composite outcome may be masked.

Drawbacks of protective ventilationThe term “protective” in the context of mechanical ventilation implies a decrease in the major components of VILI, namely atelectrauma, volutrauma and biotrauma. However, a strategy that is protective to lungs may also cause harm to other organ systems. The potential for harm caused by protective ventilation was reported in PROVHILO,86 in which patients receiving higher PEEP and lung recruitment manoeuvres developed intraoperative hypotension more frequently and needed more vasoactive drugs. These findings are at least in part in line with the finding that protective ventilation was associated with a higher incidence of intraoperative hypotension in the French trial.110

Figure 6. Settings of VT (Panel A) and PEEP (Panel B) according to observational studies of mechanical ventilation in the operation room; and settings of VT (Panel C) and PEEP (Panel D) in “non–protective” ventilation (red) and “protective” ventilation (blue) groups

Review intraoperative protective ventilation

Chap

ter

3

63

Standard of care versus unusual settings: Were the control groups of recent trials representative of clinical practice? In RCTs addressing intraoperative protective mechanical ventilation, the strategy used to treat control groups can play an important role when drawing conclusions for daily practice of general anesthesia. Metaanalyses suggest that lower tidal volumes are protective not only during long-term ventilation in critically ill patients,116,117 but also short-term ventilation during general anesthesia for surgery.117 Accordingly, anaesthesiologists have been using tidal volumes of approximately 8 to 9 mL/kg on average, and seldom higher than 10 mL/kg,74 as also illustrated in figure 6A. In contrast to this practice, the tidal volumes used in the control groups of recent RCTs were as high as 9111 to 12 mL/kg110, except to PROVHILO (figure 6B),86 which used a tidal volume of 7 mL/kg both in the intervention and in the control group. Similarly, levels of PEEP in the control arms of three out of four recent RCTs110,111,114 on protective mechanical ventilation were much lower than the standard of care at the moment the respective studies were designed (figure 6C and 6D). Taken together, these facts suggest that, among the most important recent RCTs on intraoperative protective mechanical ventilation, only the PROVHILO trial used a control group that reproduced the standard of anesthesia care at the time it was conducted. Accordingly, the PROVHILO trial addressed a major question regarding mechanical ventilation during anesthesia, namely whether the combination of high PEEP with recruitment manoeuvres confers protection against PPCs. In this study, high PEEP was not individualized, but based on previous findings from computed tomography67,88,89 and physiological studies.90,92

Intraoperative mechanical ventilation according to the utmost recent evidenceA number of reviews and commentaries have suggested that intraoperative mechanical ventilation for surgery should consist of low tidal volumes (6 to 8 mL/kg), moderate levels of PEEP (6 to 8 cmH2O), and periodic lung recruitment manoeuvres (e.g. every 30 min).5, 118-120 However, previous reviews and recommendations have been based on bundles, which do not permit to infer on the contribution of individual measures. Furthermore, the results of the largest RCT in this field (PROVHILO) could not be taken into account. Also, a recommendation regarding the use of positive pressure ventilation during induction and emergence of anesthesia, as proposed recently,119 is not supported by outcome data. Currently, the only recommendations that can be given for clinical practice are summarized in figure 7. In non-obese patients without ARDS121 undergoing open abdominal surgery, mechanical ventilation should be performed with low tidal volumes (approximately 6 to 8 mL/kg) combined with low PEEP (≤ 2 cmH2O), since higher PEEP combined with recruitment manoeuvres does not confer further protection against PPCs and can deteriorate the hemodynamics. If hypoxemia develops and provided that other causes have been excluded (e.g. hypotension, hypoventilation, pulmonary embolism etc.), the FiO2 should be increased first, followed by increase of PEEP, and recruitment manoeuvres based on stepwise increase of tidal volume during regular mechanical ventilation, according to the rescue algorithm described in the PROVHILO trial,86 provided no contra-indication is present. In patients with ARDS121 undergoing open abdominal surgery, intraoperative mechanical ventilation should be guided by the ARDS network protocol,122 whereby higher PEEP values123 may be useful in more

64

severe ARDS.124 If the target PaO2 (55 to 80 mmHg) or SpO2 (88 to 95%) cannot be achieved, a maximal lung recruitment manoeuvre with a decremental PEEP trial can be considered.125

Future perspectives

Despite the increasing number of highly qualitative RCTs on intraoperative mechanical ventilation, a number of issues remain unaddressed. While metaanalyses strongly suggest that low tidal volumes during intraoperative mechanical ventilation protect against postoperative pulmonary events, no single RCT has been able to prove this claim. Since metaanalyses in this field frequently include studies that tested intervention bundles, for example low tidal volume and high PEEP with recruitment manoeuvres versus high tidal volumes without PEEP, the estimation of the effects of single measures, for example low tidal volume or PEEP, is prone to criticism. Therefore, RCTs are most relevant for clinical practice if they test single interventions, and if control groups reproduce current standards. Whereas direct testing of the hypothesis that intraoperative low tidal volumes protect against PPCs is still lacking, ethical issues preclude such a trial.

Figure 7. Proposed settings of protective mechanical ventilation in non-obese patients during open abdominal surgery according to the concept of intraoperative permissive atelectasis

Review intraoperative protective ventilation

Chap

ter

3

65

Despite convincing evidence that PEEP and recruitment manoeuvres do not confer further protection, and may even impair hemodynamics during a ventilatory strategy based on low tidal volumes in open abdominal surgery, we do not know whether patients with obesity or undergoing one lung anesthesia procedures may benefit from those interventions. Also, we cannot rule out the possibility that an individual PEEP titration targeted on lung function could yield different results. Furthermore, it remains unclear how postoperative atelectasis, the most frequent of the different PPCs, influences the development of pulmonary infections, severe respiratory failure and affects other relevant outcome measures, including hospital length of stay and mortality. In addition, further studies should shed light on the potential contributions of ventilatory strategies during induction and emergence of anesthesia, as well as in the postoperative period (e.g. non-invasive ventilation). Accordingly, the potential of perioperative non-ventilatory measures (e.g. muscle paralysis, use of short-acting neuromuscular blocking agents as well as monitoring and reversal of muscle paralysis, early mobilization, and respiratory therapy etc.) for reducing PPCs should be investigated. Such studies are necessary to support future guidelines on the practice of perioperative mechanical ventilation and adjunctive measures in a broad spectrum of patients as well as surgical interventions, both open and laparoscopic.

Conclusions

The potential of intraoperative lung protective mechanical ventilation to reduce the incidence of PPCs is well established. RCTs have suggested that low tidal volumes, high PEEP and recruitment manoeuvres may be protective intraoperatively, but the precise role of each single intervention has been less clearly defined. A metaanalysis taking the utmost recent clinical data shows that the use of low tidal volumes, rather than PEEP, recruitment manoeuvres, or a combination of these two, is the most important determinant of protection in intraoperative mechanical ventilation. In non-obese patients without ARDS undergoing open abdominal surgery, mechanical ventilation should be performed with low tidal volumes (approximately 6 to 8 mL/kg) combined with low PEEP, since the use of higher PEEP combined with recruitment manoeuvres does not confer further protection against PPCs and can deteriorate the hemodynamics. If hypoxemia develops, and provided that other causes have been excluded, for example, hypotension, hypoventilation, pulmonary embolism etc., the FiO2 should be increased first, followed by increase of PEEP, and recruitment manoeuvres based on stepwise increase of tidal volume during regular mechanical ventilation. Further studies are warranted to guide intraoperative mechanical ventilation in a broader spectrum of patients and surgical interventions.

66

Figure LegendsFigure 1. Alterations of the extracellular matrix in lungs during mechanical ventilation and fluid administration. CS-PG, condroitin sulphate proteoglycans; HS-PG, heparan sulphate proteoglycans; ICs, inflammatory cells; IMs, inflammatory mediators; MMPs, metalloproteases; MV, mechanical ventilation; Pi, interstitial pressure; SB, spontaneous breathing; W/D, wet/dry ratio.

Figure 2. Magnetic resonance imaging (MRI) scans of lungs of three patients before and on the first day after open abdominal surgery. Images were obtained throughout spontaneous breathing and represent an average of total lung volume (TLV) during the breath cycles. Panel A – minor atelectasis; Panel B – major atelectasis; Panel C – pleural effusion. Segmentation of lungs, atelectasis (red lines) and pleura effusion (blue lines) in MRI scans was performed manually. Values were calculated for whole lungs. Note that the amounts of atelectasis and pleura effusion, two common postoperative pulmonary complications are relatively low after surgery. L, left side of chest; MRI, magnetic resonance imaging; PreOP, preoperative; PostOP, postoperative, R, right side of chest; TLV, total lung volume.

Figure 3. Effects of high and low tidal volumes (VT) at end-inspiration and end-expiration with low and high PEEP during general anesthesia. Atelectatic lung regions (red), overinflated lung regions (blue), normally aerated lung regions (white). During ventilation with low tidal volume and low PEEP, higher amounts of atelectasis are present at end-expiration and end-inspiration with minimal areas of overinflation; During ventilation with high tidal volume and low PEEP, less atelectasis is present at end-expiration and end inspiration, with increased areas of overinflation at end-inspiration. Furthermore, a higher amount of tissue collapsing and de-collapsing during breathing is present. During ventilation with low tidal volume and higher PEEP, less atelectasis is present. However, higher overinflation occurs at end- inspiration and end-expiration, with minimal collapse and reopening during breathing cycling. PEEP, positive end-expiratory pressure; VT, tidal volume.

Figure 4. Illustrative fluctuation of airway pressure during three types of lung recruitment manoeuvres for intraoperative mechanical ventilation (red lines); Panel A, “Bag squeezing” using the airway pressure-limiting valve of the anesthesia machine. The airway pressure is difficult to control, possibly resulting in overpressure, with the risk of barotrauma, or values lower than the closing pressure of small airways when controlled mechanical ventilation is resumed, with consequent lung derecruitment; Panel B, “Stepwise increase of tidal volume” during volume-controlled ventilation. PEEP is set at 12 cmH2O, the respiratory frequency at 6 to 8 breaths/min, and tidal volume increased from 8 mL/kg in steps of 4 mL/kg until the target opening pressure (e.g. 30-40 cmH2O) is achieved. After three to five breaths at that pressure, the PEEP is kept at 12 cmH2O, tidal volume reduced to 6 to 8 mL/kg, and the respiratory frequency adjusted to normocapnia; Panel C, Stepwise increase of PEEP at a constant driving pressure of 15 to 20 cmH2O in pressure-controlled ventilation. The PEEP is increased in steps of 5 cmH2O (30 to 60 s per step) up to 20 cmH2O. After three to five breaths at a PEEP level that allows achieving the target inspiratory pressure, PEEP and tidal volume are adjusted to the respective desired levels. PEEP, positive end-expiratory pressure.

Review intraoperative protective ventilation

Chap

ter

3

67

Figure 5. Odds ratios for postoperative pulmonary complications of “protective” versus “non-protective” ventilation in trials comparing different tidal volumes,85,109,113 different tidal volumes and positive end-expiratory pressure (PEEP),110-112,114 and different levels of PEEP.86

Figure 6. Settings of VT (Panel A) and PEEP (Panel B) according to observational studies of mechanical ventilation in the operation room (in Canada,128 France,129 and the USA3,74,75,130); settings of VT (Panel C) and PEEP (Panel D) in “non–protective” ventilation (red) and “protective” ventilation (blue) groups in four recent randomized controlled trials (Severgnini et al.,111 IMPROVE,110 Ge et al.,114 and PROVHILO86). PBW, predicted body weight; PEEP, positive end-expiratory pressure; VT, tidal volume.

Figure 7. Proposed settings of protective mechanical ventilation in non-obese patients during open abdominal surgery according to the concept of intraoperative permissive atelectasis. ARDS, acute respiratory distress syndrome; FIO2, inspiratory oxygen fraction of oxygen; PBW, predicted body weight; PEEP, positive end-expiratory pressure; Pplat, inspiratory airway plateau pressure; SpO2, peripheral oxygen saturation; RR, respiratory rate; VT, tidal volume.

68

References1. Canet J, Gallart L, Gomar C, Paluzie G, Valles J, Castillo J, Sabate S, Mazo V, Briones Z, Sanchis J: Prediction of

postoperative pulmonary complications in a population-based surgical cohort. Anesthesiology 2010; 113: 1338-502. Mazo V, Sabate S, Canet J, Gallart L, Gama de Abreu M, Belda J, Langeron O, Hoeft A, Pelosi P: Prospective External

Validation of a Predictive Score for Postoperative Pulmonary Complications. Anesthesiology 2014; 121: 219-313. Fernandez-Perez ER, Sprung J, Afessa B, Warner DO, Vachon CM, Schroeder DR, Brown DR, Hubmayr RD, Gajic O:

Intraoperative ventilator settings and acute lung injury after elective surgery: A nested case control study. Thorax 2009; 64: 121-7

4. Hemmes SN, Neto AS, Schultz MJ: Intraoperative ventilatory strategies to prevent postoperative pulmonary complications: A metaanalysis. Curr Opin Anaesthesiol 2013; 26: 126-33

5. Futier E, Constantin JM, Jaber S: Protective lung ventilation in operating room: A systematic review. Minerva Anestesiol 2014; 80: 726-35

6. Gajic O, Dabbagh O, Park PK, Adesanya A, Chang SY, Hou P, Anderson H, 3rd, Hoth JJ, Mikkelsen ME, Gentile NT, Gong MN, Talmor D, Bajwa E, Watkins TR, Festic E, Yilmaz M, Iscimen R, Kaufman DA, Esper AM, Sadikot R, Douglas I, Sevransky J, Malinchoc M, Illness USC, Injury Trials Group: Lung Injury Prevention Study I: Early identification of patients at risk of acute lung injury: Evaluation of lung injury prediction score in a multicenter cohort study. Am J Respir Crit Care Med 2011; 183: 462-70

7. Canet J, Gallart L: Predicting postoperative pulmonary complications in the general population. Curr Opin Anaesthesiol 2013; 26: 107-15

8. Jammer I, Wickboldt N, Sander M, Smith A, Schultz MJ, Pelosi P, Leva B, Rhodes A, Hoeft A, Walder B, Chew MS, Pearse RM: Standards for definitions and use of outcome measures for clinical effectiveness research in perioperative medicine: European Perioperative Clinical Outcome (EPCO) definitions: A statement from the ESA-ESICM joint taskforce on perioperative outcome measures. Eur J Anaesthesiol 2014; 32: 88-105

9. Arozullah AM, Daley J, Henderson WG, Khuri SF: Multifactorial risk index for predicting postoperative respiratory failure in men after major noncardiac surgery: The National Veterans Administration Surgical Quality Improvement Program. Ann Surg 2000; 232: 242-53

10. Arozullah AM, Khuri SF, Henderson WG, Daley J: Development and validation of a multifactorial risk index for predicting postoperative pneumonia after major noncardiac surgery. Ann Intern Med 2001; 135: 847-57

11. Canet J, Gallart L: Postoperative respiratory failure: Pathogenesis, prediction, and prevention. Curr Opin Crit Care 2014; 20: 56-62

12. Sabate S, Mazo V, Canet J: Predicting postoperative pulmonary complications: Implications for outcomes and costs. Curr Opin Anaesthesiol 2014; 27: 201-9

13. Smetana GW, Lawrence VA, Cornell JE: Preoperative pulmonary risk stratification for noncardiothoracic surgery: Systematic review for the American College of Physicians. Ann Intern Med 2006; 144: 581-95

14. Johnson RG, Arozullah AM, Neumayer L, Henderson WG, Hosokawa P, Khuri SF: Multivariable predictors of postoperative respiratory failure after general and vascular surgery: Results from the patient safety in surgery study. J Am Coll Surg 2007; 204: 1188-98

15. Brueckmann B, Villa-Uribe JL, Bateman BT, Grosse-Sundrup M, Hess DR, Schlett CL, Eikermann M: Development and validation of a score for prediction of postoperative respiratory complications. Anesthesiology 2013; 118: 1276-85

16. Kor DJ, Lingineni RK, Gajic O, Park PK, Blum JM, Hou PC, Hoth JJ, Anderson HL, 3rd, Bajwa EK, Bartz RR, Adesanya A, Festic E, Gong MN, Carter RE, Talmor DS: Predicting risk of postoperative lung injury in high-risk surgical patients: A multicenter cohort study. Anesthesiology 2014; 120: 1168-81

17. Higuita-Castro N, Mihai C, Hansford DJ, Ghadiali SN: Influence of airway wall compliance on epithelial cell injury and adhesion during interfacial flows. J Appl Physiol (1985) 2014; 117: 1231-42

18. Mead J, Takishima T, Leith D: Stress distribution in lungs: A model of pulmonary elasticity. J Appl Physiol 1970; 28: 596-608

19. Needham DM, Colantuoni E, Mendez-Tellez PA, Dinglas VD, Sevransky JE, Dennison Himmelfarb CR, Desai SV, Shanholtz C, Brower RG, Pronovost PJ: Lung protective mechanical ventilation and two year survival in patients with acute lung injury: Prospective cohort study. BMJ 2012; 344: e2124

20. Hager DN, Krishnan JA, Hayden DL, Brower RG, Network ACT: Tidal volume reduction in patients with acute lung injury when plateau pressures are not high. Am J Respir Crit Care Med 2005; 172: 1241-5

21. Tremblay LN, Slutsky AS: Ventilator-induced lung injury: From the bench to the bedside. Intensive Care Med 2006; 32: 24-3322. Chiumello D, Carlesso E, Cadringher P, Caironi P, Valenza F, Polli F, Tallarini F, Cozzi P, Cressoni M, Colombo A, Marini JJ,

Gattinoni L: Lung stress and strain during mechanical ventilation for acute respiratory distress syndrome. Am J Respir Crit Care Med 2008; 178: 346-55

23. Protti A, Cressoni M, Santini A, Langer T, Mietto C, Febres D, Chierichetti M, Coppola S, Conte G, Gatti S, Leopardi O, Masson S, Lombardi L, Lazzerini M, Rampoldi E, Cadringher P, Gattinoni L: Lung Stress and Strain During Mechanical

Review intraoperative protective ventilation

Chap

ter

3

69

Ventilation: Any Safe Threshold? Am J Respir Crit Care Med 2011; 183: 1354-6224. Spieth PM, Silva PL, Garcia CS, Ornellas DS, Samary CS, Moraes L, Bentes M, Morales MM, Kasper M, Guldner A, Huhle

R, Koch T, Pelosi P, de Abreu MG, Rocco PR: Modulation of Stress versus Time Product during Mechanical Ventilation Influences Inflammation as Well as Alveolar Epithelial and Endothelial Response in Rats. Anesthesiology 2015; 122: 106-16

25. Davidovich N, DiPaolo BC, Lawrence GG, Chhour P, Yehya N, Margulies SS: Cyclic stretch-induced oxidative stress increases pulmonary alveolar epithelial permeability. Am J Respir Cell Mol Biol 2013; 49: 156-64

26. Hussein O, Walters B, Stroetz R, Valencia P, McCall D, Hubmayr RD: Biophysical determinants of alveolar epithelial plasma membrane wounding associated with mechanical ventilation. Am J Physiol Lung Cell Mol Physiol 2013; 305: L478-84

27. Suki B, Hubmayr R: Epithelial and endothelial damage induced by mechanical ventilation modes. Curr Opin Crit Care 2014; 20: 17-24

28. Moriondo A, Pelosi P, Passi A, Viola M, Marcozzi C, Severgnini P, Ottani V, Quaranta M, Negrini D: Proteoglycan fragmentation and respiratory mechanics in mechanically ventilated healthy rats. J Appl Physiol 2007; 103: 747-56

29. Moriondo A, Marcozzi C, Bianchin F, Passi A, Boschetti F, Lattanzio S, Severgnini P, Pelosi P, Negrini D: Impact of respiratory pattern on lung mechanics and interstitial proteoglycans in spontaneously breathing anaesthetized healthy rats. Acta Physiol (Oxf) 2011; 203: 331-41

30. Pelosi P, Rocco PR: Effects of mechanical ventilation on the extracellular matrix. Intensive Care Med 2008; 34: 631-931. Negrini D, Passi A, Moriondo A: The role of proteoglycans in pulmonary edema development. Intensive Care Med

2008; 34: 610-832. Moriondo A, Marcozzi C, Bianchin F, Reguzzoni M, Severgnini P, Protasoni M, Raspanti M, Passi A, Pelosi P, Negrini D:

Impact of mechanical ventilation and fluid load on pulmonary glycosaminoglycans. Respir Physiol Neurobiol 2012; 181: 308-20

33. Spieth PM, Bluth T, Gama De Abreu M, Bacelis A, Goetz AE, Kiefmann R: Mechanotransduction in the lungs. Minerva Anestesiol 2014; 80: 933-41

34. Rocco PR, Dos Santos C, Pelosi P: Pathophysiology of ventilator-associated lung injury. Curr Opin Anaesthesiol 2012; 25: 123-30

35. Uhlig S: Ventilation-induced lung injury and mechanotransduction: Stretching it too far? Am J Physiol Lung Cell Mol Physiol 2002; 282: L892-6

36. Parker JC, Hernandez LA, Longenecker GL, Peevy K, Johnson W: Lung edema caused by high peak inspiratory pressures in dogs: Role of increased microvascular filtration pressure and permeability. Am Rev Respir Dis 1990; 142: 321-8

37. West JB, Tsukimoto K, Mathieu-Costello O, Prediletto R: Stress failure in pulmonary capillaries. J Appl Physiol 1991; 70: 1731-42

38. Tinsley JH, De Lanerolle P, Wilson E, Ma W, Yuan SY: Myosin light chain kinase transference induces myosin light chain activation and endothelial hyperpermeability. Am J Physiol Cell Physiol 2000; 279: C1285-9

39. Del Maschio A, Zanetti A, Corada M, Rival Y, Ruco L, Lampugnani MG, Dejana E: Polymorphonuclear leukocyte adhesion triggers the disorganization of endothelial cell-to-cell adherens junctions. J Cell Biol 1996; 135: 497-510

40. Cavanaugh KJ, Jr., Oswari J, Margulies SS: Role of stretch on tight junction structure in alveolar epithelial cells. Am J Respir Cell Mol Biol 2001; 25: 584-91

41. Egan EA: Lung inflation, lung solute permeability, and alveolar edema. J Appl Physiol Respir Environ Exerc Physiol 1982; 53: 121-5

42. Albert RK, Lakshminarayan S, Hildebrandt J, Kirk W, Butler J: Increased surface tension favors pulmonary edema formation in anesthetized dogs’ lungs. J Clin Invest 1979; 63: 1015-8

43. Pecchiari M, Monaco A, Koutsoukou A, D’Angelo E: Plasma membrane disruptions with different modes of injurious mechanical ventilation in normal rat lungs*. Crit Care Med 2012; 40: 869-75

44. D’Angelo E, Koutsoukou A, Della Valle P, Gentile G, Pecchiari M: Cytokine release, small airway injury, and parenchymal damage during mechanical ventilation in normal open-chest rats. J Appl Physiol 2008; 104: 41-9

45. Lecuona E, Saldias F, Comellas A, Ridge K, Guerrero C, Sznajder JI: Ventilator-associated lung injury decreases lung ability to clear edema in rats. Am J Respir Crit Care Med 1999; 159: 603-9

46. Planes C, Escoubet B, Blot-Chabaud M, Friedlander G, Farman N, Clerici C: Hypoxia downregulates expression and activity of epithelial sodium channels in rat alveolar epithelial cells. Am J Respir Cell Mol Biol 1997; 17: 508-18

47. Bhattacharya J, Matthay MA: Regulation and repair of the alveolar-capillary barrier in acute lung injury. Annu Rev Physiol 2013; 75: 593-615

48. Rugonyi S, Biswas SC, Hall SB: The biophysical function of pulmonary surfactant. Respir Physiol Neurobiol 2008; 163: 244-55

70

49. Vlahakis NE, Hubmayr RD: Cellular stress failure in ventilator-injured lungs. Am J Respir Crit Care Med 2005; 171: 1328-42

50. Imai Y, Parodo J, Kajikawa O, de Perrot M, Fischer S, Edwards V, Cutz E, Liu M, Keshavjee S, Martin TR, Marshall JC, Ranieri VM, Slutsky AS: Injurious mechanical ventilation and end-organ epithelial cell apoptosis and organ dysfunction in an experimental model of acute respiratory distress syndrome. JAMA 2003; 289: 2104-12

51. Rothen HU, Sporre B, Engberg G, Wegenius G, Hogman M, Hedenstierna G: Influence of gas composition on recurrence of atelectasis after a reexpansion maneuver during general anesthesia. Anesthesiology 1995; 82: 832-42

52. Joyce CJ, Williams AB: Kinetics of absorption atelectasis during anesthesia: A mathematical model. J Appl Physiol 1999; 86: 1116-25

53. D’Angelo E: Factors affecting the distribution of transpulmonary pressure in animals and in man. Bull Eur Physiopathol Respir 1984; 20: 415-22

54. Pelosi P, Ravagnan I, Giurati G, Panigada M, Bottino N, Tredici S, Eccher G, Gattinoni L: Positive end-expiratory pressure improves respiratory function in obese but not in normal subjects during anesthesia and paralysis. Anesthesiology 1999; 91: 1221-31

55. Albert RK, Hubmayr RD: The prone position eliminates compression of the lungs by the heart. Am J Respir Crit Care Med 2000; 161: 1660-5

56. Pelosi P, Goldner M, McKibben A, Adams A, Eccher G, Caironi P, Losappio S, Gattinoni L, Marini JJ: Recruitment and derecruitment during acute respiratory failure: An experimental study. Am J Respir Crit Care Med 2001; 164: 122-30

57. van Kaam AH, Lachmann RA, Herting E, De Jaegere A, van Iwaarden F, Noorduyn LA, Kok JH, Haitsma JJ, Lachmann B: Reducing atelectasis attenuates bacterial growth and translocation in experimental pneumonia. Am J Respir Crit Care Med 2004; 169: 1046-53

58. Lachmann RA, van Kaam AH, Haitsma JJ, Lachmann B: High positive end-expiratory pressure levels promote bacterial translocation in experimental pneumonia. Intensive Care Med 2007; 33: 1800-4

59. Fanelli V, Mascia L, Puntorieri V, Assenzio B, Elia V, Fornaro G, Martin EL, Bosco M, Delsedime L, Fiore T, Grasso S, Ranieri VM: Pulmonary atelectasis during low stretch ventilation: “Open lung” versus “lung rest” strategy. Crit Care Med 2009; 37: 1046-53

60. Vaneker M, Halbertsma FJ, van Egmond J, Netea MG, Dijkman HB, Snijdelaar DG, Joosten LA, van der Hoeven JG, Scheffer GJ: Mechanical ventilation in healthy mice induces reversible pulmonary and systemic cytokine elevation with preserved alveolar integrity: An in vivo model using clinical relevant ventilation settings. Anesthesiology 2007; 107: 419-26

61. Wolthuis EK, Vlaar AP, Choi G, Roelofs JJ, Juffermans NP, Schultz MJ: Mechanical ventilation using non-injurious ventilation settings causes lung injury in the absence of pre-existing lung injury in healthy mice. Crit Care 2009; 13: R1

62. Hegeman MA, Hemmes SN, Kuipers MT, Bos LD, Jongsma G, Roelofs JJ, van der Sluijs KF, Juffermans NP, Vroom MB, Schultz MJ: The extent of ventilator-induced lung injury in mice partly depends on duration of mechanical ventilation. Crit Care Res Pract 2013; 2013: 435236

63. Wellman TJ, Winkler T, Costa EL, Musch G, Harris RS, Zheng H, Venegas JG, Vidal Melo MF: Effect of local tidal lung strain on inflammation in normal and lipopolysaccharide-exposed sheep*. Crit Care Med 2014; 42: e491-500

64. Lundquist H, Hedenstierna G, Strandberg A, Tokics L, Brismar B: CT-assessment of dependent lung densities in man during general anaesthesia. Acta Radiol 1995; 36: 626-32

65. Cai H, Gong H, Zhang L, Wang Y, Tian Y: Effect of low tidal volume ventilation on atelectasis in patients during general anesthesia: A computed tomographic scan. J Clin Anesth 2007; 19: 125-9

66. Rusca M, Proietti S, Schnyder P, Frascarolo P, Hedenstierna G, Spahn DR, Magnusson L: Prevention of atelectasis formation during induction of general anesthesia. Anesth Analg 2003; 97: 1835-9

67. Neumann P, Rothen HU, Berglund JE, Valtysson J, Magnusson A, Hedenstierna G: Positive end-expiratory pressure prevents atelectasis during general anaesthesia even in the presence of a high inspired oxygen concentration. Acta Anaesthesiol Scand 1999; 43: 295-301

68. Milic-Emili J, Torchio R, D’Angelo E: Closing volume: A reappraisal (1967-2007). Eur J Appl Physiol 2007; 99: 567-8369. Juno J, Marsh HM, Knopp TJ, Rehder K: Closing capacity in awake and anesthetized-paralyzed man. J Appl Physiol 1978;

44: 238-4470. Bergman NA, Tien YK: Contribution of the closure of pulmonary units to impaired oxygenation during anesthesia.

Anesthesiology 1983; 59: 395-40171. Stanley TH, Zikria BA, Sullivan SF: The surface tension of tracheobronchial secretions during general anesthesia.

Anesthesiology 1972; 37: 445-972. Bendixen HH, Hedley-Whyte J, Laver MB: Impaired Oxygenation in Surgical Patients during General Anesthesia with

Controlled Ventilation: A Concept of Atelectasis. NEJM 1963; 269: 991-673. Esteban A, Frutos-Vivar F, Muriel A, Ferguson ND, Penuelas O, Abraira V, Raymondos K, Rios F, Nin N, Apezteguia C,

Violi DA, Thille AW, Brochard L, Gonzalez M, Villagomez AJ, Hurtado J, Davies AR, Du B, Maggiore SM, Pelosi P, Soto L, Tomicic V, D’Empaire G, Matamis D, Abroug F, Moreno RP, Soares MA, Arabi Y, Sandi F, Jibaja M, Amin P, Koh Y, Kuiper MA, Bulow HH, Zeggwagh AA, Anzueto A: Evolution of mortality over time in patients receiving mechanical ventilation.

Review intraoperative protective ventilation

Chap

ter

3

71

Am J Respir Crit Care Med 2013; 188: 220-3074. Hess DR, Kondili D, Burns E, Bittner EA, Schmidt UH: A 5-year observational study of lung-protective ventilation in the

operating room: A single-center experience. J Crit Care 2013; 28: 533 e9-1575. Levin MA, McCormick PJ, Lin HM, Hosseinian L, Fischer GW: Low intraoperative tidal volume ventilation with minimal

PEEP is associated with increased mortality. Br J Anaesth 2014; 113: 97-10876. Hong CM, Xu DZ, Lu Q, Cheng Y, Pisarenko V, Doucet D, Brown M, Aisner S, Zhang C, Deitch EA, Delphin E: Low

tidal volume and high positive end-expiratory pressure mechanical ventilation results in increased inflammation and ventilator-associated lung injury in normal lungs. Anesth Analg 2010; 110: 1652-60

77. Wrigge H, Zinserling J, Stuber F, von Spiegel T, Hering R, Wetegrove S, Hoeft A, Putensen C: Effects of mechanical ventilation on release of cytokines into systemic circulation in patients with normal pulmonary function. Anesthesiology 2000; 93: 1413-7

78. Wrigge H, Uhlig U, Zinserling J, Behrends-Callsen E, Ottersbach G, Fischer M, Uhlig S, Putensen C: The effects of different ventilatory settings on pulmonary and systemic inflammatory responses during major surgery. Anesth Analg 2004; 98: 775-81,

79. Wrigge H, Uhlig U, Baumgarten G, Menzenbach J, Zinserling J, Ernst M, Dromann D, Welz A, Uhlig S, Putensen C: Mechanical ventilation strategies and inflammatory responses to cardiac surgery: A prospective randomized clinical trial. Intensive Care Med 2005; 31: 1379-87

80. Gama de Abreu M, Heintz M, Heller AR, Sezchenyi RCM, Albrecht DM, Koch T: One-lung ventilation with high tidal volumes and zero positive end-expiratory pressure is injurious in the isolated rabbit lung model. Anesth Analg 2003; 96: 220-8

81. Kozian A, Schilling T, Schutze H, Senturk M, Hachenberg T, Hedenstierna G: Ventilatory protective strategies during thoracic surgery: Effects of alveolar recruitment maneuver and low-tidal volume ventilation on lung density distribution. Anesthesiology 2011; 114: 1025-35

82. Schilling T, Kozian A, Huth C, Buhling F, Kretzschmar M, Welte T, Hachenberg T: The pulmonary immune effects of mechanical ventilation in patients undergoing thoracic surgery. Anesth Analg 2005; 101: 957-65,

83. Michelet P, D’Journo XB, Roch A, Doddoli C, Marin V, Papazian L, Decamps I, Bregeon F, Thomas P, Auffray JP: Protective ventilation influences systemic inflammation after esophagectomy: A randomized controlled study. Anesthesiology 2006; 105: 911-9

84. Licker M, Diaper J, Villiger Y, Spiliopoulos A, Licker V, Robert J, Tschopp JM: Impact of intraoperative lung-protective interventions in patients undergoing lung cancer surgery. Crit Care 2009; 13: R41

85. Shen Y, Zhong M, Wu W, Wang H, Feng M, Tan L, Wang Q: The impact of tidal volume on pulmonary complications following minimally invasive esophagectomy: A randomized and controlled study. J Thorac Cardiovasc Surg 2013; 146: 1267-73

86. Hemmes SN, Gama de Abreu M, Pelosi P, Schultz MJ: High versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): A multicentre randomised controlled trial. Lancet 2014; 384: 495-503

87. Scott DH, Drummond GB: III. Tidal volume measurement: OK for science, but too difficult for a workstation standard? Br J Anaesth 2013; 110: 891-5

88. Brismar B, Hedenstierna G, Lundquist H, Strandberg A, Svensson L, Tokics L: Pulmonary densities during anesthesia with muscular relaxation: A proposal of atelectasis. Anesthesiology 1985; 62: 422-8

89. Reinius H, Jonsson L, Gustafsson S, Sundbom M, Duvernoy O, Pelosi P, Hedenstierna G, Freden F: Prevention of atelectasis in morbidly obese patients during general anesthesia and paralysis: A computerized tomography study. Anesthesiology 2009; 111: 979-87

90. Maisch S, Reissmann H, Fuellekrug B, Weismann D, Rutkowski T, Tusman G, Bohm SH: Compliance and dead space fraction indicate an optimal level of positive end-expiratory pressure after recruitment in anesthetized patients. Anesth Analg 2008; 106: 175-81

91. Satoh D, Kurosawa S, Kirino W, Wagatsuma T, Ejima Y, Yoshida A, Toyama H, Nagaya K: Impact of changes of positive end-expiratory pressure on functional residual capacity at low tidal volume ventilation during general anesthesia. J Anesth 2012; 26: 664-9

92. Futier E, Constantin JM, Petit A, Jung B, Kwiatkowski F, Duclos M, Jaber S, Bazin JE: Positive end-expiratory pressure improves end-expiratory lung volume but not oxygenation after induction of anaesthesia. Eur J Anaesthesiol 2010; 27: 508-13

93. Reis Miranda D, Gommers D, Struijs A, Dekker R, Mekel J, Feelders R, Lachmann B, Bogers AJ: Ventilation according to the open lung concept attenuates pulmonary inflammatory response in cardiac surgery. Eur J Cardiothorac Surg 2005; 28: 889-95

94. Cinnella G, Grasso S, Spadaro S, Rauseo M, Mirabella L, Salatto P, De Capraris A, Nappi L, Greco P, Dambrosio M: Effects of recruitment maneuver and positive end-expiratory pressure on respiratory mechanics and transpulmonary pressure during laparoscopic surgery. Anesthesiology 2013; 118: 114-22

95. Karsten J, Luepschen H, Grossherr M, Bruch HP, Leonhardt S, Gehring H, Meier T: Effect of PEEP on regional ventilation

72

during laparoscopic surgery monitored by electrical impedance tomography. Acta Anaesthesiol Scand 2011; 55: 878-8696. Zhao Z, Guttmann J, Moller K: Adaptive SLICE method: An enhanced method to determine nonlinear dynamic respiratory

system mechanics. Physiol Meas 2012; 33: 51-6497. Gattinoni L, Carlesso E, Caironi P: Stress and strain within the lung. Curr Opin Crit Care 2012; 18: 42-798. Weingarten TN, Whalen FX, Warner DO, Gajic O, Schears GJ, Snyder MR, Schroeder DR, Sprung J: Comparison of two

ventilatory strategies in elderly patients undergoing major abdominal surgery. Br J Anaesth 2010; 104: 16-2299. Wetterslev J, Hansen EG, Roikjaer O, Kanstrup IL, Heslet L: Optimizing peroperative compliance with PEEP during

upper abdominal surgery: Effects on perioperative oxygenation and complications in patients without preoperative cardiopulmonary dysfunction. Eur J Anaesthesiol 2001; 18: 358-65

100. Rothen HU, Neumann P, Berglund JE, Valtysson J, Magnusson A, Hedenstierna G: Dynamics of re-expansion of atelectasis during general anaesthesia. Br J Anaesth 1999; 82: 551-6

101. Tusman G, Groisman I, Fiolo FE, Scandurra A, Arca JM, Krumrick G, Bohm SH, Sipmann FS: Noninvasive monitoring of lung recruitment maneuvers in morbidly obese patients: The role of pulse oximetry and volumetric capnography. Anesth Analg 2014; 118: 137-44

102. Rothen HU, Sporre B, Engberg G, Wegenius G, Hedenstierna G: Re-expansion of atelectasis during general anaesthesia: A computed tomography study. Br J Anaesth 1993; 71: 788-95

103. Lin WQ, Lu XY, Cao LH, Wen LL, Bai XH, Zhong ZJ: [Effects of the lung protective ventilatory strategy on proinflammatory cytokine release during one-lung ventilation]. Ai Zheng 2008; 27: 870-3

104. Unzueta C, Tusman G, Suarez-Sipmann F, Bohm S, Moral V: Alveolar recruitment improves ventilation during thoracic surgery: A randomized controlled trial. Br J Anaesth 2012; 108: 517-24

105. Koner O, Celebi S, Balci H, Cetin G, Karaoglu K, Cakar N: Effects of protective and conventional mechanical ventilation on pulmonary function and systemic cytokine release after cardiopulmonary bypass. Intensive Care Med 2004; 30: 620-6

106. Zupancich E, Paparella D, Turani F, Munch C, Rossi A, Massaccesi S, Ranieri VM: Mechanical ventilation affects inflammatory mediators in patients undergoing cardiopulmonary bypass for cardiac surgery: A randomized clinical trial. J Thorac Cardiovasc Surg 2005; 130: 378-83

107. Wolthuis EK, Choi G, Dessing MC, Bresser P, Lutter R, Dzoljic M, van der Poll T, Vroom MB, Hollmann M, Schultz MJ: Mechanical ventilation with lower tidal volumes and positive end-expiratory pressure prevents pulmonary inflammation in patients without preexisting lung injury. Anesthesiology 2008; 108: 46-54

108. Memtsoudis SG, Bombardieri AM, Ma Y, Girardi FP: The effect of low versus high tidal volume ventilation on inflammatory markers in healthy individuals undergoing posterior spine fusion in the prone position: A randomized controlled trial. J Clin Anesth 2012; 24: 263-9

109. Treschan TA, Kaisers W, Schaefer MS, Bastin B, Schmalz U, Wania V, Eisenberger CF, Saleh A, Weiss M, Schmitz A, Kienbaum P, Sessler DI, Pannen B, Beiderlinden M: Ventilation with low tidal volumes during upper abdominal surgery does not improve postoperative lung function. Br J Anaesth 2012; 109: 263-71

110. Futier E, Constantin JM, Paugam-Burtz C, Pascal J, Eurin M, Neuschwander A, Marret E, Beaussier M, Gutton C, Lefrant JY, Allaouchiche B, Verzilli D, Leone M, De Jong A, Bazin JE, Pereira B, Jaber S, Group IS: A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. NEJM 2013; 369: 428-37

111. Severgnini P, Selmo G, Lanza C, Chiesa A, Frigerio A, Bacuzzi A, Dionigi G, Novario R, Gregoretti C, de Abreu MG, Schultz MJ, Jaber S, Futier E, Chiaranda M, Pelosi P: Protective Mechanical Ventilation during General Anesthesia for Open Abdominal Surgery Improves Postoperative Pulmonary Function. Anesthesiology 2013: 1307-21

112. Maslow AD, Stafford TS, Davignon KR, Ng T: A randomized comparison of different ventilator strategies during thoracotomy for pulmonary resection. J Thorac Cardiovasc Surg 2013; 146: 38-44

113. Sundar S, Novack V, Jervis K, Bender SP, Lerner A, Panzica P, Mahmood F, Malhotra A, Talmor D: Influence of low tidal volume ventilation on time to extubation in cardiac surgical patients. Anesthesiology 2011; 114: 1102-10

114. Ge Y, Yuan L, Jiang X, Wang X, Xu R, Ma W: [Effect of lung protection mechanical ventilation on respiratory function in the elderly undergoing spinal fusion]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2013; 38: 81-5

115. Bouadma L, Dreyfuss D, Ricard JD, Martet G, Saumon G: Mechanical ventilation and hemorrhagic shock-resuscitation interact to increase inflammatory cytokine release in rats. Crit Care Med 2007; 35: 2601-6

116. Putensen C, Theuerkauf N, Zinserling J, Wrigge H, Pelosi P: Metaanalysis: Ventilation strategies and outcomes of the acute respiratory distress syndrome and acute lung injury. Ann Intern Med 2009; 151: 566-76

117. Serpa Neto A, Cardoso SO, Manetta JA, Pereira VG, Esposito DC, Pasqualucci Mde O, Damasceno MC, Schultz MJ: Association between use of lung-protective ventilation with lower tidal volumes and clinical outcomes among patients without acute respiratory distress syndrome: A metaanalysis. JAMA 2012; 308: 1651-9

118. Goldenberg NM, Steinberg BE, Lee WL, Wijeysundera DN, Kavanagh BP: Lung-protective ventilation in the operating room: Time to implement? Anesthesiology 2014; 121: 184-8

119. Futier E, Marret E, Jaber S: Perioperative positive pressure ventilation: An integrated approach to improve pulmonary care. Anesthesiology 2014; 121: 400-8

120. Coppola S, Froio S, Chiumello D: Protective lung ventilation during general anesthesia: Is there any evidence? Crit Care 2014; 18: 210

Review intraoperative protective ventilation

Chap

ter

3

73

121. Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L, Slutsky AS: Acute respiratory distress syndrome: The Berlin Definition. JAMA 2012; 307: 2526-33

122. The Acute Respiratory Distress Syndrome N: Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. NEJM 2000; 342: 1301-8

123. Brower RG, Lanken PN, MacIntyre N, Matthay MA, Morris A, Ancukiewicz M, Schoenfeld D, Thompson BT, National Heart L, Blood Institute ACTN: Higher versus lower positive end-expiratory pressures in patients with the acute respiratory distress syndrome NEJM 2004; 351: 327-36

124. Briel M, Meade M, Mercat A, Brower RG, Talmor D, Walter SD, Slutsky AS, Pullenayegum E, Zhou Q, Cook D, Brochard L, Richard JC, Lamontagne F, Bhatnagar N, Stewart TE, Guyatt G: Higher vs lower positive end-expiratory pressure in patients with acute lung injury and acute respiratory distress syndrome: Systematic review and metaanalysis. JAMA 2010; 303: 865-73

125. Borges JB, Okamoto VN, Matos GF, Caramez MP, Arantes PR, Barros F, Souza CE, Victorino JA, Kacmarek RM, Barbas CS, Carvalho CR, Amato MB: Reversibility of lung collapse and hypoxemia in early acute respiratory distress syndrome. Am J Respir Crit Care Med 2006; 174: 268-78

126. Trillo-Alvarez C, Cartin-Ceba R, Kor DJ, Kojicic M, Kashyap R, Thakur S, Thakur L, Herasevich V, Malinchoc M, Gajic O: Acute lung injury prediction score: Derivation and validation in a population-based sample. Eur Respir J 2011; 37: 604-9

127. Kor DJ, Warner DO, Alsara A, Fernandez-Perez ER, Malinchoc M, Kashyap R, Li G, Gajic O: Derivation and Diagnostic Accuracy of the Surgical Lung Injury Prediction Model. Anesthesiology 2011; 115: 117-28

128. Lellouche F, Dionne S, Simard S, Bussières J, Dagenais F: High tidal volumes in mechanically ventilated patients increase organ dysfunction after cardiac surgery. Anesthesiology 2012; 116: 1072-82

129. Jaber S, Coisel Y, Chanques G, Futier E, Constantin JM, Michelet P, Beaussier M, Lefrant JY, Allaouchiche B, Capdevila X, Marret E: A multicentre observational study of intra-operative ventilatory management during general anaesthesia: Tidal volumes and relation to body weight. Anaesthesia 2012; 67: 999-1008

130. Chaiwat O, Vavilala MS, Philip S, Malakouti A, Neff MJ, Deem S, Treggiari MM, Wang J, Lang JD: Intraoperative adherence to a low tidal volume ventilation strategy in critically ill patients with pre-existing acute lung injury. J Crit Care 2011; 26: 144-51

Chapter 4

LAS VEGAS – Local Assessment of Ventilatory Management during General Anaesthesia for Surgery and its effects on Postoperative Pulmonary Complications: a prospective, observational, international, multicentre cohort study

Hemmes SNT, Gama de Abreu M, Pelosi P, Schultz MJ European Journal of Anaesthesiology 2013; 30(5):205-7

76

Mechanical ventilation is frequently used as a supportive strategy in intensive care medicine and anaesthesiology despite its potential to aggravate or even initiate lung injury. Indeed, overdistension of non–dependent lung regions and repetitive opening and closing of dependent lung regions may cause mechanical stress and strain eventually worsening or causing lung damage.1,2 Critically ill patients with acute respiratory distress syndrome (ARDS) requiring long–term mechanical ventilation (i.e., for days) have been found to benefit from lung–protective mechanical ventilation settings that limit airway pressures and tidal volumes.3,4 Therefore, use of such lung–protective mechanical ventilation strategies is presently recommended in these patients.5 There is growing evidence that critically ill patients at risk for but not yet suffering from lung injury also benefit from pressure– and volume–limitation during long–term mechanical ventilation.6-8 While higher levels of positive end–expiratory pressure (PEEP) with or without recruitment manoeuvres during long–term mechanical ventilation benefits selected patients with ARDS, its use is not widely recommended.4 Evidence for beneficial effects of higher PEEP levels and recruitment manoeuvres during long–term mechanical ventilation in patients with uninjured lungs is lacking.

The effects of short–term mechanical ventilation (i.e., for hours) on pulmonary integrity are still to be defined.9 The potential benefits of lung–protective mechanical ventilation strategies during general anaesthesia for surgery in patients with uninjured lungs are even questioned.10 However, results from preclinical studies of short–term mechanical ventilation in animals without lung injury suggest possible beneficial effects of pressure– and tidal volume–limitations and the use of higher PEEP levels.2,11,12 In addition, intraoperative lung-protective mechanical ventilation may attenuate postoperative lung inflammation and prevent postoperative pulmonary complications.9,13 During short–term post–operative ventilation pressure– and tidal volume–limitation may also be protective.8,14,15 Although potentially beneficial, there is insufficient evidence whether the use of higher levels of PEEP during surgery prevents postoperative pulmonary complications.16

Consensus on optimal mechanical ventilation settings during general anaesthesia for surgery is largely lacking. Knowledge on intraoperative mechanical ventilation settings is very limited. One observational study, conducted in 49 centres in France, shows 18% of patients undergoing general anaesthesia for surgery to receive tidal volumes > 10 mL/kg predicted body weight during mechanical ventilation in the operation room. In addition, 81% received mechanical ventilation without PEEP.17 Information beyond this study are scarce, though some observational studies suggest poor use of pressure– and volume–limited mechanical ventilation during general anaesthesia for surgery.9,14,18 Thus, it is unclear how widespread the concept of lung–protective mechanical ventilation is applied in other countries.

Postoperative pulmonary complications are a major contributor to postoperative complications with a reported incidence varying from 2.6 to 5.0%.19,20 Postoperative pulmonary complications can be suggested to be dependent on intra–operative mechanical ventilation settings. Considering the high number of surgical procedures performed worldwide daily, estimated to be over 600.000, and the high incidence of complications (25%) and mortality (from 3.5 to 7%) in this patient population, even a small beneficial effect of lung–protective mechanical ventilation during general anaesthesia for surgery on postoperative pulmonary complications could have significant importance.21,22

LAS VEGAS study design

Chap

ter

4

77

The LAS VEGAS study aims at characterizing current mechanical ventilation practices during general anaesthesia for surgery and assessing the dependence of postoperative pulmonary complications on intraoperative mechanical ventilation settings. As secondary endpoints, we plan to assess intraoperative complications possibly related to mechanical ventilation settings, as well as the variation of applied mechanical ventilation settings within centres and between centres on an international basis.

This large observational study will provide valuable data, which can guide optimization of intraoperative mechanical ventilation to attenuate intraoperative and postoperative pulmonary complications. Prevention of ventilator-associated lung injury (VALI) could have substantial impact on postoperative pulmonary complications, postoperative clinical course and length of hospital stay.

The LAS VEGAS study is an international observational prospective non–interventional cohort study. It will include consecutive adult patients undergoing mechanical ventilation during general anaesthesia for surgery within a 7–day study period in the first months of 2013. Patients are followed during 5 postoperative days for postoperative pulmonary complications. At day 28 after surgery length of hospital stay and in hospital mortality are recorded. Patients undergoing obstetric surgical procedures or any procedure during pregnancy, surgical procedures outside the operating room, and surgical procedures involving extra–corporal circulation are excluded from participation. Two specific patient groups will be included in the study, but analysed separately: patients undergoing one–lung ventilation during surgery, and intensive care unit patients (i.e., patients who may have already received mechanical ventilation before the surgical procedure).

Patients will be recruited in both teaching and community centres worldwide. We aim to include a minimum of 4.800 patients in at least 96 centres. Taking into account an expected minimal incidence of 2.6% postoperative pulmonary complications, we anticipate that in order to provide a sample of 120 events inclusion of at least 4.800 patients is required. This will allow for inclusion of up to 12 covariates (including but not limited to mechanical ventilation settings, fluid loading, blood transfusion, ARISCAT risk score) in a logistic regression model to analyse the effect on post-operative pulmonary complications.20 For a logistic regression analysis the number of events divided by the number of predictor variables should be at least ten.23

The data to be collected are all collected as part of routine clinical care. Predefined risk factors for postoperative pulmonary complications (including, but not restricted to physical status, smoking status, chronic co-morbidity, transfusion of red blood cells, urgency of surgery, surgical procedure, fluid loading, use of epidural anaesthesia), intraoperative mechanical ventilation settings (ventilatory mode, airway pressures, tidal volume size, PEEP, respiratory rate), intraoperative complications possibly related to the mechanical ventilation strategy (oxygen desaturation, need for unplanned recruitment manoeuvres, need for pressure reduction, need for expiratory flow limitation, hypotension, need for vasoactive drugs, new arrhythmias) and postoperative pulmonary complications (new or prolonged invasive or non–invasive mechanical ventilation, need for oxygen therapy, respiratory failure, pneumonia, acute respiratory distress syndrome, pneumothorax) are recorded from the medical chart.19,20,24 In patients who are admitted to the intensive care unit after surgery a more detailed follow–up is performed. These patients will be part of an elective substudy, which can provide important information on post–operative critical care.

78

All data is collected on paper case record forms, unless a local electronic system to register intraoperative and/or postoperative data is used (e.g., a patient data management system). All collected data are transcribed by local investigators into a web–based electronic Case Report Form (open source software OpenClinica).

In this multicenter study we will check for design effects. Design effects measures the effect of clustering due to multi–site recruitment of subjects. Student’s t–test or Mann-Whitney U–tests will be used to compare continuous variables and chi-squared tests for categorical variables. Comparison between and within groups will be performed using one–way ANOVA and post–hoc analyses for continuous variables. Study parameters of both patients receiving mechanical ventilation prior to operation and of patients undergoing one–lung ventilation during surgery will be analysed separately. Data from the intensive care unit sub–study will be analysed separately. To identify potential factors associated with intra– and post–operative pulmonary complications univariate analyses will be performed. A multivariate logistic regression model will be used to identify independent risk factors for post-operative pulmonary complications. To enter new terms into the model a stepwise approach will be used, with a limit of p < 0.05 to enter the terms. Time to event variables will be analysed using Cox regression and visualized by Kaplan–Meier curves. Statistical significance will be considered at p < 0.05.

All participating centres will submit the study protocol to their local Institutional Review Board for ethical judgment and obtain document of proof that the study has been subjected to ethical review and granted approval/favourable opinion. If required, ethical approval must be obtained before the start of inclusion.

The study is registered at Clinicaltrials.gov with registration identifier NCT 01601223.

The study will be performed by the LAS VEGAS collaboration on behalf of the European Society of Anaesthesiology (ESA). National coordinators will identify and recruit local participating centres, translate all study documents and ensure that all local necessary ethical and regulatory approvals are obtained before start of patient inclusion. They will assist, train and monitor local centres, ensuring conduction of the study according to the Good Clinical Practice guidelines of the International Conference on Harmonisation (ICH–GCP).25 Local coordinators in individual participating centres will provide scientific and structural leadership in their centre. They will guarantee the integrity of data collection and ensure timely completion of the dataset.

After publication of the primary results in a peer–reviewed medical journal, on request the pooled dataset will be available for all members of the LAS VEGAS collaboration for secondary analyses after judgment and approval of scientific quality and validity by the Steering Committee.

Funding

The study is funded by the European Society of Anaesthesiology (ESA) and supported by infrastructure provided by the Society.

LAS VEGAS study design

Chap

ter

4

79

References1. Slutsky A. Lung injury caused by mechanical ventilation. Chest 1999; 116: 9S-15S2. Tremblay L and Slutsky A. Ventilator-induced lung injury: from the bench to the bedside. Intensive care medicine 2006;

32: 24-333. The ARDS Network. Acute respiratory distress syndrome: The Berlin definition. JAMA 2012; 307: 2526-25334. Putensen C, Theuerkauf N, Zinserling J, Wrigge H and Pelosi P. Metaanalysis: ventilation strategies and outcomes of

the acute respiratory distress syndrome and acute lung injury. Annals of internal medicine 2009; 151: 566-5765. Dellinger R, Levy M, Carlet J, et al. Surviving Sepsis Campaign: international guidelines for management of severe

sepsis and septic shock: 2008. Critical Care Medicine 2008; 36: 296-3276. Gajic O, Dara S, Mendez J, et al. Ventilator-associated lung injury in patients without acute lung injury at the onset of

mechanical ventilation. Critical Care Medicine 2004; 32: 1817-18247. Gajic O, Frutos-Vivar F, Esteban A, Hubmayr R and Anzueto A. Ventilator settings as a risk factor for acute respiratory

distress syndrome in mechanically ventilated patients. Intensive Care Medicine 2005; 31: 922-9268. Determann R, Royakkers A, Wolthuis E, et al. Ventilation with lower tidal volumes as compared with conventional tidal

volumes for patients without acute lung injury: a preventive randomized controlled trial. Critical Care 2008; 14: R19. Fernandez-Perez E, Keegan M, Brown D, Hubmayr R and Gajic O. Intraoperative tidal volume as a risk factor for

respiratory failure after pneumonectomy. Anesthesiology 2006; 105: 14-1810. Wrigge H, Uhlig U, Zinserling J, et al. The effects of different ventilatory settings on pulmonary and systemic inflammatory

responses during major surgery. Anesthesia and Analgesia 2004; 98: 775-78111. Wolthuis E, Vlaar A, Choi G, Roelofs J, Juffermans N and Schultz M. Mechanical ventilation using non-injurious ventilation

settings causes lung injury in the absence of pre-existing lung injury in healthy mice. Critical Care 2009; 13: R112. Moriondo A, Marcozzi C, Bianchin F, et al. Impact of mechanical ventilation and fluid load on pulmonary

glycosaminoglycans. Respir Physiol Neurobiology 2012; 181: 308-32013. Schultz M. Lung-protective mechanical ventilation with lower tidal volumes in patients not suffering from acute lung

injury: a review of clinical studies. Med Sci Monit 2008; 14: RA22-2614. Lellouche F, Dionne S, Simard S, Bussieres J and Dagenais F. High tidal volumes in mechanically ventilated patients

increase organ dysfunction after cardiac surgery. Anesthesiology 2012; 116: 1072-108215. Pelosi P and Gama de Abreu M. Tidal volumes during general anesthesia: size does matter! Anesthesiology 2012; 116:

985-98616. Imberger G, McIlroy D, Pace NL, Wetterslev J, Brok J and Moller AM. Positive end-expiratory pressure (PEEP) during

anaesthesia for the prevention of mortality and postoperative pulmonary complications. Cochrane Database Syst Rev 2010: CD007922

17. Jaber S, Coisel Y, Chanques G, et al. A multicentre observational study of intra-operative ventilatory management during general anaesthesia: tidal volumes and relation to body weight. Anaesthesia 2012; 67: 999-1008

18. Blum JM, Maile M, Park PK, et al. A Description of Intraoperative Ventilator Management in Patients with Acute Lung Injury and the Use of Lung Protective Ventilation Strategies. Anesthesiology 2010; 115: 75-82

19. Smetana GW, Lawrence VA and Cornell JE. Preoperative pulmonary risk stratification for noncardiothoracic surgery: systematic review for the American College of Physicians. Annals of Internal Medicine 2006; 144: 581-595

20. Canet J, Gallart L, Gomar C, et al. Prediction of Postoperative Pulmonary Complications in a Population-based Surgical Cohort. Anesthesiology 2010; 113: 1338-1350

21. Weiser TG, Regenbogen SE, Thompson KD, et al. An estimation of the global volume of surgery: a modelling strategy based on available data. Lancet 2008; 372: 139-144

22. Ghaferi AA, Birkmeyer JD and Dimick JB. Variation in hospital mortality associated with inpatient surgery. NEJM 2009; 361: 1368-1375

23. Bagley SC, White H and Golomb BA. Logistic regression in the medical literature: standards for use and reporting, with particular attention to one medical domain. Journal of clinical epidemiology 2001; 54: 979-985

24. Arozullah AM, Khuri SF, Henderson WG and Daley J. Development and validation of a multifactorial risk index for predicting postoperative pneumonia after major noncardiac surgery. Annals of Internal Medicine 2001; 135: 847-857

25. International conference on harmonisation of technical requirements for registration of pharmaceuticals for human use. ICH Harmonised Tripartite Guideline: Guideline for Good Clinical Practice. Journal of postgraduate medicine 2001; 47: 199-203

Chapter 5

Intraoperative Ventilation Strategies and Patient Outcomes Following Surgery: an International Observational Study (LAS VEGAS)

Hemmes SNT, Gama de Abreu M, Pelosi P, Schultz MJ for the The LAS VEGAS Investigators for PROVE Network*, and the Clinical Trial Network of the European Society of Anaesthesiology *PROVE Network: the PROtective VEntilation Network Manuscript submitted

82

Abstract

Background. Postoperative pulmonary complications (PPCs) increase morbidity and mortality of surgical patients. The intraoperative ventilation strategy may affect occurrence of PPCs.

Objective. To describe practice of intraoperative ventilation and to determine associations between ventilator settings and occurrence of PPCs.

Design, data source, setting and patients. Prospective observational study in adult patients requiring invasive ventilation during general anaesthesia for surgery in 2013.

Measurements. The primary outcome measure was development of PPCs in the first five postoperative days. A multivariable model was built to quantify the net effect of intraoperative ventilation characteristics on the occurrence of PPCs, while controlling for other perioperative risk factors.

Results. Data collection was complete in 8,241 patients (146 hospitals in 29 countries); PPCs occurred in 861 (10.4%) patients, arising most commonly within three days after surgery. Median tidal volume was 500.0 (454.2–550.5) mL resulting in 8.1 (7.2–9.1) mL/kg predicted body weight. The positive end–expiratory pressure (PEEP) level was 4.0 (0.0–5.0) cm H2O. PEEP levels of 0 and 5 cm H2O were used most frequently. Recruitment manoeuvres were performed in < 10% of patients. Increased levels of PEEP were independently associated with the occurrence of PPCs (odds ratio 1.06 (95% CI) 1.01–1.12; p = 0.020)).

Limitations. The observational character of this study precludes causal inferences.

Conclusion. Low tidal volumes are frequently used, but PEEP levels above 5 cm H2O and recruitment manoeuvres are seldom applied. Occurrence of PPCs is independently associated with the use of increasing intraoperative PEEP levels.

The study is registered at Clinicaltrials.gov, number NCT01601223.

LAS VEGAS study

Chap

ter

5

83

Introduction

Mechanical ventilation is often considered a harmless intervention during general anaesthesia for surgery. However, ventilation can induce overdistension as well as repetitive opening and closing of lung units, resulting in ventilator–induced lung injury and even the acute respiratory distress syndrome (ARDS).1 Several randomized controlled trials showed that ventilation with low tidal volumes (VT) is associated with improved outcome in patients with ARDS and in critically ill patients without ARDS,2–4 and this knowledge has profoundly changed ventilation practice in intensive care units.5–7

Theoretically, surgery patients with uninjured lungs could also benefit from intraoperative ventilation with low VT.3 Three randomized controlled trials in patients planned for major abdominal surgery showed that intraoperative ventilation with low VT and positive end–expiratory pressures (PEEP) with recruitment manoeuvres reduced occurrence of postoperative pulmonary complications (PPCs).8-10 One metaanalysis, however, suggested that VT, rather than PEEP, is the major factor responsible for lung protection during intraoperative ventilation.11 One recent multicentre audit in Australia showed that surgery patients remain to receive high VT (median 9.5 (8.5–10.4 mL/kg predicted body weight) and higher levels of PEEP (median 5.0 (4.0–5.0) cm H2O).12 Another study showed that the average size of VT during intraoperative ventilation is 7.8 ± 1.5 mL/kg predicted body weight, and that > 60% of patients receive ≥ 5 cm H2O of PEEP.13 Thus while there is controversy regarding VT, PEEP levels have increased, despite the fact that PEEP may not protect against PPC, and maybe even cause harm.11,14

We conducted the ‘Local ASsessment of VEntilatory management during General Anesthesia for Surgery, LAS VEGAS’ study, an international multicentre prospective observational study, to describe current intraoperative ventilation practice, and to determine associations between ventilator settings and patient outcome. We hypothesized that VT < 10 mL/kg PBW and PEEP levels ≥ 5 cm H2O were commonly used and both to be associated with a reduced occurrence of PPCs.

Methods

Study design and sites The LAS VEGAS study was co–sponsored and endorsed by the European Society of Anaesthesiology (ESA), which assisted in developing the electronic case record forms, and hosted the electronic database, without influencing study design, conduct, data analysis, and final reporting. The Writing Committee drafted the manuscript, and the Steering Committee provided revisions and comments. The study was registered at Clinicaltrials.gov (NCT01601223).

Study sites were recruited through the Clinical Trial Network of the ESA. Sites sought approval from the respective Institutional Review Board (research ethics committee), and if required, obtained written informed consent from individual patients. National coordinators assisted local coordinators and monitored the study according to the ‘International Conference on

84

Harmonization (Good Clinical Practice) guidelines.15 Local coordinators arranged regulatory approvals, monitored local researchers, and ensured integrity and timely completion of data collection.

Study population and data collectionAdult patients receiving invasive ventilation during general anaesthesia for elective or non–elective surgery were eligible for participation in the study, which ran for seven predefined days in each country, selected by the national coordinator, in the period between January 14th and March 4th, 2013. Patients were excluded from participation if they were aged < 18 years, or scheduled for pregnancy related surgery, surgical procedures outside the operating room, or procedures involving cardio-pulmonary bypass. Data from patients undergoing thoracic surgery or one–lung ventilation during surgery, and those who had received ventilation at any time in the previous 30 days were collected, but excluded from the present analysis. Centres performing more than 180 surgical procedures per week were allowed to randomly select either 25% or 50% of their eligible patients for inclusion (ALEA Version 2.2 | AMC/ALEA). The randomization procedure is described in the Supplement.

Baseline characteristics and preoperative risk factors for PPCs were identified from previous studies.16–19 During the intraoperative period we collected data describing ventilation settings and vital parameters hourly, and scored complications possibly related to ventilation, including episodes of hypoxia, use of recruitment manoeuvres, airway pressure reduction, presence of expiratory flow limitation, hypotension, use of vasoactive drugs, and new arrhythmias. Postoperative residual curarisation with neuromuscular blocking agents (NMBAs), defined as train–of–four stimulation (TOF) ratio < 0.9 was documented.

OutcomesThe following PPCs were scored daily from the day of surgery until hospital discharge or postoperative day 5: 1) need for supplementary oxygen (due to PaO2 < 60 mmHg or SpO2 < 90% in room air, excluding oxygen supplementation given as standard care or as continuation of preoperative therapy), 2) respiratory failure (PaO2 < 60 mmHg or SpO2 < 90% despite oxygen therapy, or need for non-invasive mechanical ventilation), 3) unplanned new or prolonged invasive or non–invasive mechanical ventilation, 4) acute respiratory distress syndrome, 5) pneumonia, and 6) pneumothorax. Length of hospital stay and in–hospital mortality was censored at postoperative day 28. Definitions of intraoperative complications, possible types of recruitment manoeuvres, and postoperative complications are provided in eTables 1, 2 and 3. Patient data were anonymized before entry onto a password secured, web–based electronic case record form (OpenClinica, Boston, MA, USA).

Statistical analysisSince the reported incidences of PPCs vary between 2.6% and 5.0%,16,17 we anticipated that to provide a sample of at least 120 PPC events, enrolment of 4,800 or more patients was required. This would allow up to 12 covariates in a logistic regression model for predicting PPCs.20

Part of the statistical analysis plan was published previously.21 Patients with incomplete data in main intraoperative ventilation settings including VT size (VT expressed in mL, and in mL/kg,

LAS VEGAS study

Chap

ter

5

85

PBW), PEEP and peak pressure level, or oxygen fraction in inspired air (FiO2) were censored. The ventilation settings were examined for plausibility and excluded if: VT < 100 or > 1500 mL, VT < 2 or > 20 mL/kg PBW, PEEP > 30 cm H2O, peak pressure < 5 or > 70 cm H2O, and FiO2 < 0.21 or > 1.0. The remaining cohort included only complete data of main ventilation settings and the primary endpoint.

Patient demographics, surgical characteristics, intraoperative characteristics, intraoperative complications, PPCs, length of hospital stay and in–hospital mortality are presented using descriptive statistics. Demographic data are presented as medians (with the 25 and 75% quartiles). Hourly–collected intraoperative variables were averaged per patient and are presented as medians (with the 25 and 75% quartiles). The occurrence of PPCs is presented as a collapsed composite of PPCs in the first five postoperative days. We calculated Kaplan-Meier estimates of survival curves, and we used log–rank tests to compare survival distributions in the overall cohort and also in the subgroups of BMI (< 35 vs. ≥ 35 kg/m2), ARISCAT (< 26 vs. ≥ 26) and laparoscopic vs. non–laparoscopic surgery. We considered PPC development as any PPC within five postoperative days, and within one patient. We censored data used for Kaplan–Meier estimates when patients did not have a PPC during the study period, or when patients were lost to follow–up before the end of postoperative day 5.

A multivariable model was built to quantify the association between intraoperative ventilation characteristics, including VT (expressed in mL/kg PBW), PEEP level, peak pressure, and FiO2, and the occurrence of PPCs, while controlling for other known preoperative and intraoperative risk factors for PPCs.15–18. We conducted multi–level analyses to adjust for clustering of the data. Therefore, generalized linear mixed model was used to determine predictors of PPC, by modelling PPC as the dependent variable over time. Independent variables were selected according to biologic plausibility, and when a p value less than 0.2 was found in the univariate analysis. Then, the generalized linear mixed model was conducted with these selected predictor variables as fixed effects, and centres where participants were treated (cluster) as a random effect. Effects were expressed as an average odds ratio (OR) with their respective 95% confidence interval (95% CI). The OR represents how the predictor affects outcome for the combined population of all clusters instead of one specific cluster. In the multivariable model statistical significance was set at a p value < 0.05.

Propensity scores for PPCs were estimated for each patient with logistic regression using 20 relevant baseline, intraoperative and postoperative characteristics (age, sex, BMI, ARISCAT,19 preoperative SpO2, functional status, smoking, chronic obstructive pulmonary disease (COPD), chronic comorbidity, respiratory infection < 30 days, preoperative anaemia, type of surgery, planned duration of surgery, condition of surgery, type of incision, fluid intake, epidural anaesthesia, use of and reversal of NMBAs, and postoperative residual curarisation) and correcting for the clustering of the data. The propensity score reflects the propensity in the range of 0 to 1 to present PPCs given a set of known variables, and is an attempt to adjust for potential selection bias, confounding factors, and differences between groups. Patients with missing data were excluded from the database. Based on the propensity score weighted estimators for the clustered data we constructed a propensity score–matched cohort. Matching was performed using nearest neighbour matching without replacement, with each patient with PPCs matched to

86

Figure 1. Data collection and selection of cohort for analysisIRB: institutional review board; VT: tidal volume; mL: millilitres; kg: kilogram; PBW: predicted body weight; PEEP: positive end-expiratory pressure; Ppeak: peak pressure; FiO2: inspired fraction of oxygen

LAS VEGAS study

Chap

ter

5

87

three patients without PPCs. A calibre width of 0.15 of the standard deviation of the logit of the propensity score was used for the development of matching. Absolute standardized differences were computed to evaluate matching effectiveness.22

A PROBIT regression analysis was used to characterize the dose–response relationship between the intraoperative PEEP level and the probability of PPCs. A cubic term was used in the final model for PEEP. The cubic term was chosen, because we hypothesized that the relationship between PEEP and PPCs is curvilinear with low probability in low levels, some plateau, and high probability in high levels. This was confirmed by the inspection of the residuals.

Post–hoc analyses were conducted for patients with body mass index (BMI) < versus ≥ 35 kg/m2, for patients undergoing laparoscopic versus non–laparoscopic surgery, and patients with low versus high risk for PPCs, according to the Assess Respiratory Risk in Surgical Patients in Catalonia risk (ARISCAT) score (< versus ≥ 26, respectively).19 The chi square test for categorical variables and Mann–Whitney U test for continuous variables were performed to compare the intraoperative ventilation strategies, intraoperative complications and PPCs within each subgroup. Statistical significance was considered to be at p < 0.05. All analyses were performed with R version 3.1 (http://www.R-project.org/).

Role of funding sourceThe European Society of Anaesthesiology supported LAS VEGAS. The funding sources had no role in study design, data analysis or interpretation, or writing of the manuscript.

Results

Two hundred and nineteen centres in 38 countries expressed interest in participating in the LAS VEGAS study. Of them, 73 in seven countries did not obtain formal approval from the local Institutional Review Board, or had other reasons not to participate (figure 1). One centre used the randomization program to reduce the number of patients to 50%, and another one to 25% of eligible patients. The list of participating centres, countries and respective numbers of included patients is presented in eTable 4; hospital characteristics of participating centres are given in eTable 5. In total 10,523 patients were recruited of whom 656 patients were excluded due to ventilation preceding surgery, or thoracic surgery, and 1,540 patients had missing data in main ventilation settings; in 86 patients the postoperative follow–up data was missed, yielding 8,241 patients for the analysis (figure 1). In the BMI subgroups an additional 18 patients were excluded due to missing weight and/or height. Of 7,594 patients follow–up was complete until the day of hospital discharge. Demographics and surgical characteristics of patients are shown in table 1 and eTable 6.

Intraoperative ventilation characteristicsAnaesthetists preferred volume– over pressure–controlled ventilation. The pressure support mode and combined modes of ventilation were seldom used (table 2). The most frequently chosen VT was 500 mL, or 7 to 9 mL/kg PBW, with limited variation (figure 2, table 2). VT was

88

lower than 8 and higher than 10 mL/kg PBW in 46% and 12% of patients, respectively (figure 2). The most frequently applied PEEP levels were 0 and 5 cm H2O (table 2, figure 2). PEEP levels were lower than 2, 2 to 5 and ≥ 6 cm H2O in 30%, 61%, and 9% of patients, respectively. Recruitment manoeuvres were applied in 9.5% of all patients.

Patient outcomesThe most frequent intraoperative complication was hypotension (table 3). PPCs occurred in 861 patients (10.4%), arising most commonly within three days after surgery (figure 3). The need for supplemental oxygen arose in 700 patients (8.5%), and 138 patients developed respiratory

Figure 2. Distribution of intraoperative ventilation settingsTidal volume size (expressed in mL and in mL/kg predicted body weight, PBW), level of PEEP (cm H2O), and peak pressure (cm H2O) presented for the whole group; patients with body mass index (BMI) lower than 35 kg/m2 versus equal or higher than 35 kg/m2; patients undergoing laparoscopic versus non–laparoscopic surgery; and patients with a low versus high risk for PPCs, according to the ARISCAT score (lower versus equal and higher than 26)

LAS VEGAS study

Chap

ter

5

89

failure (1.7%). ARDS, pneumonia, and pneumothorax occurred in less than 1% of patients (table 4). Patients who developed PPCs experienced a longer stay in hospital and greater mortality (eFigure 1 and 2).

The occurrence of PPCs was associated with increasing PEEP and peak pressure levels, and not associated with VT (table 5 and eTable 7). PPCs were also associated with patient age above 50 years, preoperative oxygen saturation ≤ 90%, current smoking, recent respiratory infection, and preoperative anaemia. PPCs were associated with the following surgery characteristics: urgent or emergency surgery, planned duration of surgery longer than two hours, abdominal incision, intraoperative events (fluid administration of 2,000 mL or more, hypoxemia, use of vasoactive drugs), postoperative residual curarisation, and no reversal of neuromuscular blockade (table 5 and eTable 7). The categorization of PEEP and tidal volume in tertiles did not change the results (eTable 8).

The characteristics of the propensity score matched cohort are show in eTable 9. Patients developing PPCs experienced more intraoperative complications, including hypoxemia, hypotension, arrhythmias, and need for vasoactive drugs (eTable 9). In the propensity score matched cohort, PEEP levels and peak pressure were associated with the development of PPCs (table 6).

Figure 4 and eFigure 4 respectively show the association between levels of PEEP and peak pressure with the occurrence of PPCs.

Post–hoc analysesIn patients with BMI ≥ 35 kg/m2, VT was higher (table 2, figure 2, eFigure 3). Also, FiO2, peak airway pressures and PEEP levels were higher (table 2) and recruitment manoeuvres more often used. The most frequent intraoperative complication was hypotension and need for vaso–active drugs, followed by de–saturation.

In laparoscopic vs. non–laparoscopic surgery, and in patients with ARISCAT scores19 ≥ 26 vs. < 26, VT sizes were comparable, but peak airway pressures as well as levels of PEEP were higher, and recruitment manoeuvres more frequently applied (table 3). FiO2 was higher in laparoscopic surgery, and lower in patients with ARISCAT ≥ 26.

The occurrence of PPCs was higher in patients with a BMI ≥ 35 kg/m2, and those with an ARISCAT score ≥ 26 (table 4).

In patients with BMI ≥ 35 kg/m2, higher PEEP levels and peak pressures were independently associated with the occurrence of PPCs (table 5). Also in patients with ARISCAT score ≥ 26, higher PEEP levels were independently associated with the occurrence of PPCs (table 5). Age > 50 years, urgent or emergency surgery, planned duration of surgery longer than two hours, intraoperative de–saturation, postoperative residual curarisation, and no reversal of neuromuscular blockade were consistently associated with PPCs (table 5 and eTables 10, 11, 12).

90

Tabl

e 1.

Dem

ogra

phic

s an

d su

rgic

al c

hara

cter

istic

s

Varia

ble

All p

atien

tsBM

I < 3

5BM

I ≥ 3

5N

on la

paro

scop

icLa

paro

scop

icAR

ISCA

T <

26AR

ISCA

T ≥

26

Mal

e se

x45

.0 (3

711/

8241

)46

.4 (4

020/

7507

)30

.4 (2

18/7

16)

47.6

(315

7/66

36)

34.5

(554

/160

5)43

.7 (2

664/

6097

)48

.8 (1

047/

2144

)

Age

(yea

rs)

54.0

(40.

0 –

66.0

)54

.0 (4

0.0

– 66

.0)

54.0

(43.

0 –

63.0

)54

.0 (4

1.0

– 67

.0)

50.0

(37.

0 –

63.0

)50

.0 (3

7.0

– 63

.0)

63.0

(51.

0 –

72.0

)

≤ 50

44.1

(363

5/82

39)

44.2

(332

1/75

06)

43.1

(308

/715

)42

.3 (2

806/

6634

)51

.7 (8

29/1

605)

51.5

(313

7/60

96)

23.2

(498

/214

3)

51 –

80

52.2

(430

4/82

39)

52.0

(390

3/75

06)

55.0

(393

/715

)53

.8 (3

571/

6634

)45

.7 (7

33/1

605)

46.7

(284

9/60

96)

67.9

(145

5/21

43)

> 80

3.6

(300

/823

9)3.

8 (2

82/7

506)

2.0

(14/

715)

3.9

(257

/663

4)2.

7 (4

3/16

05)

1.8

(110

/609

6)8.

9 (1

90/2

143)

BMI (

kg/m

2 )26

.2 (2

3.4

– 30

.0)

25.8

(23.

0 –

28.8

)38

.3 (3

6.2

– 41

.4)

26.1

(23.

3 –

29.7

)26

.9 (2

3.5

– 31

.2)

26.1

(23.

2 –

29.7

)26

.8 (2

3.7

– 30

.5)

Unde

rwei

ght (

< 18

.5)

2.4

(194

/822

3)2.

6 (1

94/7

507)

---2.

3 (1

51/6

623)

2.7

(43/

1600

)2.

3 (1

38/6

085)

2.6

(56/

2138

)

No o

besit

y (1

8.5

– 24

.9)

37.1

(305

1/82

23)

40.6

(305

1/75

07)

---37

.8 (2

506/

6623

)34

.1 (5

45/1

600)

38.6

(234

7/60

85)

32.9

(704

/213

8)

Ove

rwei

ght

(≥ 2

5.0

- 29.

9)35

.5 (2

917/

8223

)38

.9 (2

917/

7507

)---

36.2

(240

0/66

23)

32.3

(517

/160

0)35

.1 (2

133/

6085

)36

.7 (7

84/2

138)

Obe

sity

Clas

s 1

(30.

0 to

34.

9)16

.3 (1

345/

8223

)17

.9 (1

345/

7507

)---

16.0

(106

1/66

23)

17.8

(284

/160

0)16

.0 (9

76/6

085)

17.3

(369

/213

8)

Obe

sity

Clas

s 2

(35.

0 to

39.

9)5.

7 (4

64/8

223)

---64

.8 (4

64/7

16)

5.3

(351

/662

3)7.

1 (1

13/1

600)

5.4

(329

/608

5)6.

3 (1

35/2

138)

Mor

bid

obes

ity (≥

40)

3.1

(252

/822

3)---

35.2

(252

/716

)2.

3 (1

54/6

623)

6.1

(98/

1600

)2.

7 (1

62/6

085)

4.2

(90/

2138

)

ASA

phys

ical

stat

us c

lass

ifica

tion

syst

em

AS

A 1

29.5

(242

6/82

29)

31.5

(236

0/74

97)

8.7

(62/

714)

29.0

(192

1/66

29)

31.6

(505

/160

0)35

.8 (2

177/

6089

)11

.6 (2

49/2

140)

AS

A 2

49.2

(405

1/82

29)

48.5

(363

5/74

97)

57.6

(411

/714

)48

.5 (3

214/

6629

)52

.3 (8

37/1

600)

49.6

(301

9/60

89)

48.2

(103

2/21

40)

AS

A 3

19.6

(161

5/82

29)

18.5

(138

4/74

97)

31.5

(225

/714

)20

.5 (1

362/

6629

)15

.8 (2

53/1

600)

13.9

(844

/608

9)36

.0 (7

71/2

140)

AS

A 4

1.6

(134

/822

9)1.

5 (1

16/7

497)

2.1

(15/

714)

1.9

(129

/662

9)0.

3 (5

/160

0)0.

8 (4

7/60

89)

4.1

(87/

2140

)

AS

A 5

0.0

(3/8

229)

0.0

(2/7

497)

0.1

(1/7

14)

0.0

(3/6

629)

0.0

(0/1

600)

0.0

(2/6

089)

0.0

(1/2

140)

Func

tiona

l sta

tus

Non

dep

ende

nt93

.0 (7

662/

8237

)93

.2(6

994/

7503

)91

.3 (6

54/7

16)

92.3

(612

2/66

33)

96.0

(154

0/16

04)

94.8

(577

8/60

94)

87.9

(188

4/21

43)

Par

tially

dep

ende

nt5.

9 (4

83/8

237)

5.7

(428

/750

3)7.

3 (5

2/71

6)6.

5 (4

29/6

633)

3.4

(54/

1604

)4.

2 (2

58/6

094)

10.5

(225

/214

3)

Tot

ally

dep

ende

nt1.

1 (9

2/82

37)

1.1

(81/

7503

)1.

4 (1

0/71

6)1.

2 (8

2/66

33)

0.6

(10/

1604

)1.

0 (5

8/60

94)

1.6

(34/

2143

)

Bloo

d tr

ansf

usio

n (<

24h

pr

eope

rativ

ely)

0.

6 (5

2/82

41)

0.7

(49/

7507

)0.

4 (3

/716

)0.

7 (4

7/66

36)

0.3

(5/1

605)

0.1

(8/6

097)

2.1

(44/

2144

)

ARIS

CAT

scor

e$15

.0 (3

.0 –

26.

0)15

.0 (3

.0 –

26.

0)16

.0 (3

.0 –

27.

0)11

.0 (3

.0 –

23.

0)18

.0 (1

5.0

– 31

.0)

3.0

(0.0

– 1

6.0)

34.0

(30.

0 –

41.0

)

< 26

74.0

(609

7/82

41)

74.5

(559

4/75

07)

68.6

(491

/716

)76

.6 (5

084/

6636

)63

.1 (1

013/

1605

)10

0.0

(609

7/60

97)

---

26 –

44

21.8

(179

3/82

41)

21.3

(159

6/75

07)

26.7

(191

/716

)19

.3 (1

281/

6636

)31

.9 (5

12/1

605)

---83

.6 (1

793/

2144

)

> 44

4.3

(351

/824

1)4.

2 (3

17/7

507)

4.7

(34/

716)

4.1

(271

/663

6)5.

0 (8

0/16

05)

---16

.4 (3

51/2

144)

Preo

pera

tive

SpO

2 (%

)98

.0 (9

6.0

– 99

.0)

98.0

(96.

0 –

99.0

)97

.0 (9

5.0

– 98

.0)

98.0

(96.

0 –

99.0

)98

.0 (9

6.0

– 99

.0)

98.0

(97.

0 –

99.0

)97

.0 (9

5.0

– 98

.0)

≥ 96

83.6

(611

2/73

11)

84.5

(562

5/66

57)

74.4

(476

/640

)82

.8 (4

860/

5871

)86

.9 (1

252/

1440

)89

.9 (4

850/

5397

)65

.9 (1

262/

1914

)

91–9

515

.2 (1

114/

7311

)14

.4 (9

60/6

657)

23.6

(151

/640

)15

.9 (9

36/5

871)

12.4

(178

/144

0)10

.1 (5

47/5

397)

29.6

(567

/191

4)

≤ 9

01.

2 (8

5/73

11)

1.1

(72/

6657

)2.

0 (1

3/64

0)1.

3 (7

5/58

71)

0.7

(10/

1440

)0.

0 (0

/539

7)4.

4 (8

5/19

14)

Haem

oglo

bin

– g/

dL*

13.8

(12.

6 –

14.9

)13

.8 (1

2.6

– 14

.9)

13.8

(12.

8 –

14.9

)13

.8 (1

2.6

– 14

.9)

13.7

(12.

8 –

14.6

)13

.9 (1

2.9

– 15

.0)

13.3

(11.

6 –

14.5

)Pr

eope

rativ

e an

aem

ia

(Hb

≤ 10

g/d

l)3.

4 (2

38/6

929)

3.5

(224

/633

2)2.

2 (1

3/58

0)3.

7 (2

06/5

542)

2.3

(32/

1387

)1.

0 (5

0/49

27)

9.4

(188

/200

2)

LAS VEGAS study

Chap

ter

5

91

Tabl

e 1.

Dem

ogra

phic

s an

d su

rgic

al c

hara

cter

istic

s

Varia

ble

All p

atien

tsBM

I < 3

5BM

I ≥ 3

5N

on la

paro

scop

icLa

paro

scop

icAR

ISCA

T <

26AR

ISCA

T ≥

26

Mal

e se

x45

.0 (3

711/

8241

)46

.4 (4

020/

7507

)30

.4 (2

18/7

16)

47.6

(315

7/66

36)

34.5

(554

/160

5)43

.7 (2

664/

6097

)48

.8 (1

047/

2144

)

Age

(yea

rs)

54.0

(40.

0 –

66.0

)54

.0 (4

0.0

– 66

.0)

54.0

(43.

0 –

63.0

)54

.0 (4

1.0

– 67

.0)

50.0

(37.

0 –

63.0

)50

.0 (3

7.0

– 63

.0)

63.0

(51.

0 –

72.0

)

≤ 50

44.1

(363

5/82

39)

44.2

(332

1/75

06)

43.1

(308

/715

)42

.3 (2

806/

6634

)51

.7 (8

29/1

605)

51.5

(313

7/60

96)

23.2

(498

/214

3)

51 –

80

52.2

(430

4/82

39)

52.0

(390

3/75

06)

55.0

(393

/715

)53

.8 (3

571/

6634

)45

.7 (7

33/1

605)

46.7

(284

9/60

96)

67.9

(145

5/21

43)

> 80

3.6

(300

/823

9)3.

8 (2

82/7

506)

2.0

(14/

715)

3.9

(257

/663

4)2.

7 (4

3/16

05)

1.8

(110

/609

6)8.

9 (1

90/2

143)

BMI (

kg/m

2 )26

.2 (2

3.4

– 30

.0)

25.8

(23.

0 –

28.8

)38

.3 (3

6.2

– 41

.4)

26.1

(23.

3 –

29.7

)26

.9 (2

3.5

– 31

.2)

26.1

(23.

2 –

29.7

)26

.8 (2

3.7

– 30

.5)

Unde

rwei

ght (

< 18

.5)

2.4

(194

/822

3)2.

6 (1

94/7

507)

---2.

3 (1

51/6

623)

2.7

(43/

1600

)2.

3 (1

38/6

085)

2.6

(56/

2138

)

No o

besit

y (1

8.5

– 24

.9)

37.1

(305

1/82

23)

40.6

(305

1/75

07)

---37

.8 (2

506/

6623

)34

.1 (5

45/1

600)

38.6

(234

7/60

85)

32.9

(704

/213

8)

Ove

rwei

ght

(≥ 2

5.0

- 29.

9)35

.5 (2

917/

8223

)38

.9 (2

917/

7507

)---

36.2

(240

0/66

23)

32.3

(517

/160

0)35

.1 (2

133/

6085

)36

.7 (7

84/2

138)

Obe

sity

Clas

s 1

(30.

0 to

34.

9)16

.3 (1

345/

8223

)17

.9 (1

345/

7507

)---

16.0

(106

1/66

23)

17.8

(284

/160

0)16

.0 (9

76/6

085)

17.3

(369

/213

8)

Obe

sity

Clas

s 2

(35.

0 to

39.

9)5.

7 (4

64/8

223)

---64

.8 (4

64/7

16)

5.3

(351

/662

3)7.

1 (1

13/1

600)

5.4

(329

/608

5)6.

3 (1

35/2

138)

Mor

bid

obes

ity (≥

40)

3.1

(252

/822

3)---

35.2

(252

/716

)2.

3 (1

54/6

623)

6.1

(98/

1600

)2.

7 (1

62/6

085)

4.2

(90/

2138

)

ASA

phys

ical

stat

us c

lass

ifica

tion

syst

em

AS

A 1

29.5

(242

6/82

29)

31.5

(236

0/74

97)

8.7

(62/

714)

29.0

(192

1/66

29)

31.6

(505

/160

0)35

.8 (2

177/

6089

)11

.6 (2

49/2

140)

AS

A 2

49.2

(405

1/82

29)

48.5

(363

5/74

97)

57.6

(411

/714

)48

.5 (3

214/

6629

)52

.3 (8

37/1

600)

49.6

(301

9/60

89)

48.2

(103

2/21

40)

AS

A 3

19.6

(161

5/82

29)

18.5

(138

4/74

97)

31.5

(225

/714

)20

.5 (1

362/

6629

)15

.8 (2

53/1

600)

13.9

(844

/608

9)36

.0 (7

71/2

140)

AS

A 4

1.6

(134

/822

9)1.

5 (1

16/7

497)

2.1

(15/

714)

1.9

(129

/662

9)0.

3 (5

/160

0)0.

8 (4

7/60

89)

4.1

(87/

2140

)

AS

A 5

0.0

(3/8

229)

0.0

(2/7

497)

0.1

(1/7

14)

0.0

(3/6

629)

0.0

(0/1

600)

0.0

(2/6

089)

0.0

(1/2

140)

Func

tiona

l sta

tus

Non

dep

ende

nt93

.0 (7

662/

8237

)93

.2(6

994/

7503

)91

.3 (6

54/7

16)

92.3

(612

2/66

33)

96.0

(154

0/16

04)

94.8

(577

8/60

94)

87.9

(188

4/21

43)

Par

tially

dep

ende

nt5.

9 (4

83/8

237)

5.7

(428

/750

3)7.

3 (5

2/71

6)6.

5 (4

29/6

633)

3.4

(54/

1604

)4.

2 (2

58/6

094)

10.5

(225

/214

3)

Tot

ally

dep

ende

nt1.

1 (9

2/82

37)

1.1

(81/

7503

)1.

4 (1

0/71

6)1.

2 (8

2/66

33)

0.6

(10/

1604

)1.

0 (5

8/60

94)

1.6

(34/

2143

)

Bloo

d tr

ansf

usio

n (<

24h

pr

eope

rativ

ely)

0.

6 (5

2/82

41)

0.7

(49/

7507

)0.

4 (3

/716

)0.

7 (4

7/66

36)

0.3

(5/1

605)

0.1

(8/6

097)

2.1

(44/

2144

)

ARIS

CAT

scor

e$15

.0 (3

.0 –

26.

0)15

.0 (3

.0 –

26.

0)16

.0 (3

.0 –

27.

0)11

.0 (3

.0 –

23.

0)18

.0 (1

5.0

– 31

.0)

3.0

(0.0

– 1

6.0)

34.0

(30.

0 –

41.0

)

< 26

74.0

(609

7/82

41)

74.5

(559

4/75

07)

68.6

(491

/716

)76

.6 (5

084/

6636

)63

.1 (1

013/

1605

)10

0.0

(609

7/60

97)

---

26 –

44

21.8

(179

3/82

41)

21.3

(159

6/75

07)

26.7

(191

/716

)19

.3 (1

281/

6636

)31

.9 (5

12/1

605)

---83

.6 (1

793/

2144

)

> 44

4.3

(351

/824

1)4.

2 (3

17/7

507)

4.7

(34/

716)

4.1

(271

/663

6)5.

0 (8

0/16

05)

---16

.4 (3

51/2

144)

Preo

pera

tive

SpO

2 (%

)98

.0 (9

6.0

– 99

.0)

98.0

(96.

0 –

99.0

)97

.0 (9

5.0

– 98

.0)

98.0

(96.

0 –

99.0

)98

.0 (9

6.0

– 99

.0)

98.0

(97.

0 –

99.0

)97

.0 (9

5.0

– 98

.0)

≥ 96

83.6

(611

2/73

11)

84.5

(562

5/66

57)

74.4

(476

/640

)82

.8 (4

860/

5871

)86

.9 (1

252/

1440

)89

.9 (4

850/

5397

)65

.9 (1

262/

1914

)

91–9

515

.2 (1

114/

7311

)14

.4 (9

60/6

657)

23.6

(151

/640

)15

.9 (9

36/5

871)

12.4

(178

/144

0)10

.1 (5

47/5

397)

29.6

(567

/191

4)

≤ 9

01.

2 (8

5/73

11)

1.1

(72/

6657

)2.

0 (1

3/64

0)1.

3 (7

5/58

71)

0.7

(10/

1440

)0.

0 (0

/539

7)4.

4 (8

5/19

14)

Haem

oglo

bin

– g/

dL*

13.8

(12.

6 –

14.9

)13

.8 (1

2.6

– 14

.9)

13.8

(12.

8 –

14.9

)13

.8 (1

2.6

– 14

.9)

13.7

(12.

8 –

14.6

)13

.9 (1

2.9

– 15

.0)

13.3

(11.

6 –

14.5

)Pr

eope

rativ

e an

aem

ia

(Hb

≤ 10

g/d

l)3.

4 (2

38/6

929)

3.5

(224

/633

2)2.

2 (1

3/58

0)3.

7 (2

06/5

542)

2.3

(32/

1387

)1.

0 (5

0/49

27)

9.4

(188

/200

2)

92

Varia

ble

All p

atien

tsBM

I < 3

5 BM

I ≥ 3

5N

on la

paro

scop

icLa

paro

scop

icAR

ISCA

T <

26AR

ISCA

T ≥

26

Chro

nic

co-m

orbi

dity

- a

patie

nt ca

n ha

ve m

ore

than

one

co-m

orbi

dity

Met

asta

tic ca

ncer

4.1

(340

/824

1)4.

3 (3

24/7

507)

2.2

(16/

716)

4.4

(294

/663

6)2.

9 (4

6/16

05)

1.8

(111

/609

7)10

.7 (2

29/2

144)

Chro

nic

kidn

ey

dysf

uncti

on3.

2 (2

60/8

241)

3.2

(243

/750

7)2.

2 (1

6/71

6)3.

5 (2

31/6

636)

1.8

(29/

1605

)2.

0 (1

23/6

097)

6.4

(137

/214

4)

COPD

6.1

(502

/824

1)6.

0 (4

50/7

507)

7.3

(52/

716)

6.2

(413

/663

6)5.

5 (8

9/16

05)

4.8

(292

/609

7)9.

8 (2

10/2

144)

Hear

t fai

lure

5.8

(482

/824

1)5.

8 (4

33/7

507)

6.6

(47/

716)

6.1

(404

/663

6)4.

9 (7

8/16

05)

4.7

(289

/609

7)9.

0 (1

93/2

144)

Obs

truc

tive

sleep

ap

noea

2.2

(181

/824

1)1.

2 (9

2/75

07)

12.4

(89/

716)

2.0

(135

/663

6)2.

9 (4

6/16

05)

2.0

(123

/609

7)2.

7 (5

8/21

44)

Neur

omus

cula

r di

seas

e**

1.0

(80/

8241

)1.

0 (7

3/75

07)

1.0

(7/7

16)

1.1

(72/

6636

)0.

5 (8

/160

5)1.

0 (6

1/60

97)

0.9

(19/

2144

)

Live

r dys

func

tion

1.0

(79/

8241

)1.

0 (7

4/75

07)

0.7

(5/7

16)

1.1

(70/

6636

)0.

6 (9

/160

5)0.

7 (4

4/60

97)

1.6

(35/

2144

)

Surg

ical

pro

cedu

re -

a pa

tient

can

have

mor

e th

an o

ne ty

pe o

f sur

gica

l pro

cedu

re

Low

er g

astr

o-in

testi

nal

10.5

(868

/824

1)10

.9 (8

15/7

507)

6.8

(49/

716)

9.4

(621

/663

6)15

.4 (2

47/1

605)

6.3

(386

/609

7)22

.5 (4

82/2

144)

Uppe

r GI,

hepa

to-b

iliar

y, pa

ncre

as

13.8

(113

8/82

41)

12.8

(959

/750

7)24

.7 (1

77/7

16)

6.5

(431

/663

6)44

.0 (7

06/1

605)

10.6

(649

/609

7)22

.8 (4

89/2

144)

Vasc

ular

surg

ery§

3.1

(257

/824

1)3.

2 (2

40/7

507)

2.4

(17/

716)

3.9

(257

/663

6)0.

0 (0

/160

5)2.

9 (1

75/6

097)

3.8

(82/

2144

)

Aorti

c su

rger

y0.

7 (5

9/82

41)

0.7

(54/

7507

)0.

7 (5

/716

)0.

9 (5

9/66

36)

0.0

(0/1

605)

0.3

(17/

6097

)2.

0 (4

2/21

44)

Neur

osur

gery

, hea

d &

ne

ck20

.2 (1

666/

8241

)20

.7 (1

553/

7507

)15

.4 (1

10/7

16)

25.0

(166

1/66

36)

0.3

(5/1

605)

23.9

(145

5/60

97)

9.8

(211

/214

4)

Urol

ogic

al a

nd k

idne

y8.

9 (7

32/8

241)

9.1

(680

/750

7)7.

1 (5

1/71

6)9.

0 (5

99/6

636)

8.3

(133

/160

5)6.

9 (4

22/6

097)

14.5

(310

/214

4)

Gyna

ecol

ogic

al

11.7

(963

/824

1)11

.6 (8

69/7

507)

12.7

(91/

716)

8.1

(537

/663

6)26

.5 (4

26/1

605)

10.9

(665

/609

7)13

.9 (2

98/2

144)

Endo

crin

e su

rger

y2.

0 (1

64/8

241)

2.0

(147

/750

7)2.

4 (1

7/71

6)2.

3 (1

53/6

636)

0.7

(11/

1605

)2.

3 (1

41/6

097)

1.1

(23/

2144

)

Tran

spla

nt0.

4 (3

2/82

41)

0.4

(32/

7507

)0.

0 (0

//71

6)0.

5 (3

0/66

36)

0.1

(2/1

605)

0.1

(5/6

097)

1.3

(27/

2144

)

Plas

tic, c

utan

eous

, bre

ast

10.8

(891

/824

1)10

.9 (8

20/7

507)

9.9

(71/

716)

13.3

(880

/663

6)0.

7 (1

1/16

05)

13.3

(810

/609

7)3.

8 (8

1/21

44)

Bone

, joi

nt, t

raum

a,

spin

e16

.2 (1

337/

8241

)16

.3 (1

220/

7507

)15

.6 (1

12/7

16)

19.8

(131

6/66

36)

1.3

(21/

1605

)18

.7 (1

141/

6097

)9.

1 (1

96/2

144)

Oth

er p

roce

dure

6.1

(500

/824

1)6.

2 (4

64/7

507)

5.0

(36/

716)

6.7

(442

/663

6)3.

6 (5

8/16

05)

7.2

(436

/609

7)3.

0 (6

4/21

44)

Surg

ical

tech

niqu

e - a

pati

ent c

an h

ave

mor

e th

an o

ne ty

pe o

f sur

gica

l pro

cedu

re

Ope

n ab

dom

inal

surg

ery

17.4

(143

4/82

41)

17.5

(131

0/75

07)

16.9

(121

/716

)21

.4 (1

420/

6636

)---

7.0

(424

/609

7)47

.1 (1

010/

2144

)

Lapa

rosc

opic

surg

ery

17.9

(147

8/82

41)

17.0

(127

7/75

07)

27.4

(196

/716

)---

93.0

(149

3/16

05)

15.6

(953

/609

7)24

.5 (5

25/2

144)

Lapa

rosc

opic

ass

isted

su

rger

y1.

8 (1

45/8

241)

1.7

(127

/750

7)2.

5 (1

8/71

6)---

9.0

(144

/160

5)1.

0 (6

2/60

97)

3.9

(83/

2144

)

Perip

hera

l sur

gery

18.7

(154

0/82

41)

18.6

(140

0/75

07)

18.7

(134

/716

)23

.2 (1

538/

6636

)0.

1 (2

/160

5)22

.2 (1

351/

6097

)8.

8 (1

89/2

144)

Oth

er44

.9 (3

702/

8241

)45

.9 (3

447/

7507

)35

.1 (2

51/7

16)

55.7

(369

8/66

36)

0.2

(4/1

605)

54.5

(332

0/60

97)

17.8

(382

/214

4)

Urg

ency

of s

urge

ry#

Ele

ctive

90.8

(748

6/82

40)

90.7

(680

9/75

06)

93.2

(667

/716

)91

.1 (6

047/

6635

)89

.7 (1

439/

1605

)92

.3 (5

629/

6096

)86

.6 (1

857/

2144

)

Urg

ent

7.2

(595

/824

0)7.

3 (5

45/7

506)

5.9

(42/

716)

7.0

(466

/663

5)8.

0 (1

29/1

605)

6.4

(393

/609

6)9.

4 (2

02/2

144)

Em

erge

ncy

1.9

(159

/824

0)2.

0 (1

52/7

506)

1.0

(7/7

16)

1.8

(122

/663

5)2.

3 (3

7/16

05)

1.2

(74/

6096

)4.

0 (8

5/21

44)

LAS VEGAS study

Chap

ter

5

93

Varia

ble

All p

atien

tsBM

I < 3

5 BM

I ≥ 3

5N

on la

paro

scop

icLa

paro

scop

icAR

ISCA

T <

26AR

ISCA

T ≥

26

Chro

nic

co-m

orbi

dity

- a

patie

nt ca

n ha

ve m

ore

than

one

co-m

orbi

dity

Met

asta

tic ca

ncer

4.1

(340

/824

1)4.

3 (3

24/7

507)

2.2

(16/

716)

4.4

(294

/663

6)2.

9 (4

6/16

05)

1.8

(111

/609

7)10

.7 (2

29/2

144)

Chro

nic

kidn

ey

dysf

uncti

on3.

2 (2

60/8

241)

3.2

(243

/750

7)2.

2 (1

6/71

6)3.

5 (2

31/6

636)

1.8

(29/

1605

)2.

0 (1

23/6

097)

6.4

(137

/214

4)

COPD

6.1

(502

/824

1)6.

0 (4

50/7

507)

7.3

(52/

716)

6.2

(413

/663

6)5.

5 (8

9/16

05)

4.8

(292

/609

7)9.

8 (2

10/2

144)

Hear

t fai

lure

5.8

(482

/824

1)5.

8 (4

33/7

507)

6.6

(47/

716)

6.1

(404

/663

6)4.

9 (7

8/16

05)

4.7

(289

/609

7)9.

0 (1

93/2

144)

Obs

truc

tive

sleep

ap

noea

2.2

(181

/824

1)1.

2 (9

2/75

07)

12.4

(89/

716)

2.0

(135

/663

6)2.

9 (4

6/16

05)

2.0

(123

/609

7)2.

7 (5

8/21

44)

Neur

omus

cula

r di

seas

e**

1.0

(80/

8241

)1.

0 (7

3/75

07)

1.0

(7/7

16)

1.1

(72/

6636

)0.

5 (8

/160

5)1.

0 (6

1/60

97)

0.9

(19/

2144

)

Live

r dys

func

tion

1.0

(79/

8241

)1.

0 (7

4/75

07)

0.7

(5/7

16)

1.1

(70/

6636

)0.

6 (9

/160

5)0.

7 (4

4/60

97)

1.6

(35/

2144

)

Surg

ical

pro

cedu

re -

a pa

tient

can

have

mor

e th

an o

ne ty

pe o

f sur

gica

l pro

cedu

re

Low

er g

astr

o-in

testi

nal

10.5

(868

/824

1)10

.9 (8

15/7

507)

6.8

(49/

716)

9.4

(621

/663

6)15

.4 (2

47/1

605)

6.3

(386

/609

7)22

.5 (4

82/2

144)

Uppe

r GI,

hepa

to-b

iliar

y, pa

ncre

as

13.8

(113

8/82

41)

12.8

(959

/750

7)24

.7 (1

77/7

16)

6.5

(431

/663

6)44

.0 (7

06/1

605)

10.6

(649

/609

7)22

.8 (4

89/2

144)

Vasc

ular

surg

ery§

3.1

(257

/824

1)3.

2 (2

40/7

507)

2.4

(17/

716)

3.9

(257

/663

6)0.

0 (0

/160

5)2.

9 (1

75/6

097)

3.8

(82/

2144

)

Aorti

c su

rger

y0.

7 (5

9/82

41)

0.7

(54/

7507

)0.

7 (5

/716

)0.

9 (5

9/66

36)

0.0

(0/1

605)

0.3

(17/

6097

)2.

0 (4

2/21

44)

Neur

osur

gery

, hea

d &

ne

ck20

.2 (1

666/

8241

)20

.7 (1

553/

7507

)15

.4 (1

10/7

16)

25.0

(166

1/66

36)

0.3

(5/1

605)

23.9

(145

5/60

97)

9.8

(211

/214

4)

Urol

ogic

al a

nd k

idne

y8.

9 (7

32/8

241)

9.1

(680

/750

7)7.

1 (5

1/71

6)9.

0 (5

99/6

636)

8.3

(133

/160

5)6.

9 (4

22/6

097)

14.5

(310

/214

4)

Gyna

ecol

ogic

al

11.7

(963

/824

1)11

.6 (8

69/7

507)

12.7

(91/

716)

8.1

(537

/663

6)26

.5 (4

26/1

605)

10.9

(665

/609

7)13

.9 (2

98/2

144)

Endo

crin

e su

rger

y2.

0 (1

64/8

241)

2.0

(147

/750

7)2.

4 (1

7/71

6)2.

3 (1

53/6

636)

0.7

(11/

1605

)2.

3 (1

41/6

097)

1.1

(23/

2144

)

Tran

spla

nt0.

4 (3

2/82

41)

0.4

(32/

7507

)0.

0 (0

//71

6)0.

5 (3

0/66

36)

0.1

(2/1

605)

0.1

(5/6

097)

1.3

(27/

2144

)

Plas

tic, c

utan

eous

, bre

ast

10.8

(891

/824

1)10

.9 (8

20/7

507)

9.9

(71/

716)

13.3

(880

/663

6)0.

7 (1

1/16

05)

13.3

(810

/609

7)3.

8 (8

1/21

44)

Bone

, joi

nt, t

raum

a,

spin

e16

.2 (1

337/

8241

)16

.3 (1

220/

7507

)15

.6 (1

12/7

16)

19.8

(131

6/66

36)

1.3

(21/

1605

)18

.7 (1

141/

6097

)9.

1 (1

96/2

144)

Oth

er p

roce

dure

6.1

(500

/824

1)6.

2 (4

64/7

507)

5.0

(36/

716)

6.7

(442

/663

6)3.

6 (5

8/16

05)

7.2

(436

/609

7)3.

0 (6

4/21

44)

Surg

ical

tech

niqu

e - a

pati

ent c

an h

ave

mor

e th

an o

ne ty

pe o

f sur

gica

l pro

cedu

re

Ope

n ab

dom

inal

surg

ery

17.4

(143

4/82

41)

17.5

(131

0/75

07)

16.9

(121

/716

)21

.4 (1

420/

6636

)---

7.0

(424

/609

7)47

.1 (1

010/

2144

)

Lapa

rosc

opic

surg

ery

17.9

(147

8/82

41)

17.0

(127

7/75

07)

27.4

(196

/716

)---

93.0

(149

3/16

05)

15.6

(953

/609

7)24

.5 (5

25/2

144)

Lapa

rosc

opic

ass

isted

su

rger

y1.

8 (1

45/8

241)

1.7

(127

/750

7)2.

5 (1

8/71

6)---

9.0

(144

/160

5)1.

0 (6

2/60

97)

3.9

(83/

2144

)

Perip

hera

l sur

gery

18.7

(154

0/82

41)

18.6

(140

0/75

07)

18.7

(134

/716

)23

.2 (1

538/

6636

)0.

1 (2

/160

5)22

.2 (1

351/

6097

)8.

8 (1

89/2

144)

Oth

er44

.9 (3

702/

8241

)45

.9 (3

447/

7507

)35

.1 (2

51/7

16)

55.7

(369

8/66

36)

0.2

(4/1

605)

54.5

(332

0/60

97)

17.8

(382

/214

4)

Urg

ency

of s

urge

ry#

Ele

ctive

90.8

(748

6/82

40)

90.7

(680

9/75

06)

93.2

(667

/716

)91

.1 (6

047/

6635

)89

.7 (1

439/

1605

)92

.3 (5

629/

6096

)86

.6 (1

857/

2144

)

Urg

ent

7.2

(595

/824

0)7.

3 (5

45/7

506)

5.9

(42/

716)

7.0

(466

/663

5)8.

0 (1

29/1

605)

6.4

(393

/609

6)9.

4 (2

02/2

144)

Em

erge

ncy

1.9

(159

/824

0)2.

0 (1

52/7

506)

1.0

(7/7

16)

1.8

(122

/663

5)2.

3 (3

7/16

05)

1.2

(74/

6096

)4.

0 (8

5/21

44)

94

Varia

ble

All p

atien

tsBM

I < 3

5BM

I ≥ 3

5N

on la

paro

scop

icLa

paro

scop

icAR

ISCA

T <

26AR

ISCA

T ≥

26

Plan

ned

dura

tion

of su

rger

y

≤ 2

hou

rs69

.5 (5

724/

8233

)69

.9 (5

242/

7499

)65

.6 (4

70/7

16)

68.3

(452

5/66

28)

74.7

(119

9/16

05)

87.0

(529

6/60

89)

20.0

(428

/214

4)

> 2

– 3

hou

rs19

.2 (1

584/

8233

)18

.8 (1

410/

7499

)23

.6 (1

69/7

16)

20.0

(132

6/66

28)

16.1

(258

/160

5)10

.8 (6

55/6

089)

43.3

(929

/214

4)

> 3

hou

rs11

.2 (9

25/8

233)

11.3

(847

/749

9)10

.8 (7

7/71

6)11

.7 (7

77/6

628)

9.2

(148

/160

5)2.

3 (1

38/6

089)

36.7

(787

/214

4)

Dura

tion

of su

rger

y (m

in)†

73.0

(41.

0 –

125.

0)71

.0 (4

0.0

– 12

5.0)

80.0

(50.

0 –

130.

0)75

.0 (4

0.0

– 12

6.0)

68.0

(45.

0 –

118.

0)60

.0 (3

5.0

– 93

.0)

146.

5 (9

4.0

– 21

0.0)

Dura

tion

of a

naes

thes

ia

(min

)††

104.

0 (6

7.0

– 16

5.5)

101.

0 (6

5.0

– 16

3.0)

113.

0 (7

5.0

– 17

1.0)

105.

0 (6

7.0

– 16

5.0)

96.0

(69.

0 –

155.

0)87

.0 (6

0.0

– 12

5.0)

190.

0 (1

25.0

260.

0)

Data

is p

rese

nted

as:

med

ian

(QR)

or p

ropo

rtion

(n/N

); BM

I: Bo

dy M

ass

Inde

x; A

SA: A

mer

ican

Soc

iety

of A

nest

hesi

olog

y; $

ARSI

CAT

scor

e <

26 p

redi

cts

a lo

w ri

sk fo

r pos

tope

rativ

e pu

lmon

ary

com

plic

ation

s (PP

Cs);

ARSI

CAT

scor

e ≥

26 p

redi

cts a

n in

term

edia

te to

hig

h ris

k fo

r PPC

s; S

pO2:

Per

iphe

ral O

xyge

n Sa

tura

tion;

*La

bora

tory

val

ues:

hae

mog

lobi

n w

as c

olle

cted

w

hen

avai

labl

e fr

om c

olle

ction

with

in ro

utine

car

e; C

OPD

: Chr

onic

Obs

truc

tive

Pulm

onar

y Di

seas

e; *

*Neu

rom

uscu

lar d

isea

se a

ffecti

ng th

e re

spira

tory

sys

tem

; §Va

scul

ar s

urge

ry is

ca

rotid

end

arte

rect

omy,

aor

tic su

rger

y an

d pe

riphe

ral v

ascu

lar t

aken

toge

ther

; #U

rgen

cy o

f sur

gery

: ele

ctive

: sur

gery

that

is sc

hedu

led

in a

dvan

ce b

ecau

se it

doe

s not

invo

lve

a m

edic

al

emer

genc

y, u

rgen

t: su

rger

y re

quire

d w

ithin

< 4

8 ho

urs,

em

erge

ncy:

non

-ele

ctive

surg

ery

perf

orm

ed w

hen

the

patie

nt’s

life

or w

ell-b

eing

is in

dire

ct je

opar

dy; †

Dura

tion

of su

rger

y is

the

time

betw

een

skin

inci

sion

and

clos

ure

of th

e in

cisio

n. †

†Dur

ation

of a

naes

thes

ia is

the

time

betw

een

star

t ind

uctio

n an

d ex

tuba

tion

or d

ischa

rge

from

ope

ratio

n ro

om if

mec

hani

cal

venti

latio

n re

mai

ned

LAS VEGAS study

Chap

ter

5

95

Discussion

The main findings of the LAS VEGAS study were that: 1) median tidal volume (VT) was 500.0 (454.2 – 550.5) mL or 8.1 (7.2 – 9.1) mL/kg PBW, PEEP level was 4.0 (0.0 – 5.0) cm H2O and recruitment manoeuvres were rarely performed; 2) PPCs occurred frequently within 3 days after surgery and were associated with longer length of hospital stay and mortality; 3) increasing levels of PEEP but not VT size was independently associated with PPCs.

LAS VEGAS is the largest investigation of the association between intraoperative ventilation strategies and PPCs conducted to date. We investigated both preoperative patient–related and procedure–related risk factors combined with intraoperative complications that were identified in previous investigations.1,16–19,23 The prospective design of the study not only improved the completeness of the data collection, but also avoided any effect of time. The international character makes it representative for many countries.

The most frequently chosen VT and respiratory rate represent the default settings on many anaesthesia ventilators, suggesting a lack of individualization. However, VT size was lower than in previous publications on intraoperative ventilation,24–26 implying that VT size has decreased, at least in the countries that participated in this investigation. Our findings further suggest that there is uncertainty on what PEEP level to use during surgery (0 or 5 cm H2O), but that PEEP levels higher than 5 cm H2O are almost never used, even not in patients with a high BMI, or with high risk for PPCs.27,28

In our cohort, PPCs occurred at a higher rate than previously reported.1,19 The most frequent pulmonary complication was need for oxygen excluding supplementation given as standard of care or continuation of preoperative therapy. When this PPC is not considered, the incidence of PPCs was 1.9%. Patients who developed PPCs had a longer stay in hospital and higher mortality. The most frequent intraoperative complication in all groups was hypotension and need for vasoactive drugs, followed by desaturation, mainly in patients with a BMI ≥ 35 kg/m2 and an ARISCAT score ≥ 26. This observation might reflect attempts to reverse alveolar collapse and atelectasis and their hemodynamic side effects.

The results of the multivariable model seem to be in contrast with results from previous investigations. While several randomized controlled trials showed that the use of low VT is associated with improved outcome of intensive care unit patients,3 and a recent metaanalysis suggested that VT is the major factor responsible for lung protection during intraoperative ventilation,11 the present analysis shows that the occurrence of PPCs is not associated with VT size. Also, differently from the results of three recently published randomized controlled trials,8-10 we found that use of level of PEEP ≥ 7 cmH2O is associated with an increased incidence of PPCs. Both in obese and in high risk patients, increasing PEEP was also independently associated with development of PPCs. There are two possible explanations for these discrepancies. First, in patients included in the present cohort, VT size was remarkably lower than in previous investigations in the intensive care unit,6,7 and in the operation room.3,11,30 Indeed, variations in VT were much smaller, and VT was seldom > 10 ml/kg PBW. Consequently, an association between VT size and occurrence of PPCs would likely need a much larger group of patients.

96

Table 2. Intraoperative ventilation characteristics

Variable All patients BMI < 35 BMI ≥ 35 p value* Non-laparoscopic Laparoscopic p value* ARISCAT < 26 ARISCAT ≥ 26 p value*

Ventilation mode

Volume Control 69.8 (5683/8147) 70.1 (5204/7421) 65.9 (467/709)

0.041

69.0 (4525/6558) 72.9 (1158/1589)

0.005

68.9 (4154/6026) 72.1 (1529/2121)

< 0.0001Pressure Control 17.0 (1383/8147) 16.8 (1250/7421) 18.3 (130/709) 17.3 (1132/6558) 15.8 (251/1589) 17.9 (1081/6026) 14.2 (302/2121)Pressure Support or Spontaneous 5.9 (482/8147) 5.8 (431/7421) 7.1 (50/709) 6.3 (412/6558) 4.4 (70/1589) 6.2 (373/6026) 5.2 (109/2121)

Other$ 7.4 (599/8147) 7.2 (536/7421) 8.7 (62/709) 7.5 (489/6558) 6.9 (110/1589) 6.9 (418/6026) 8.5 (181/2121)Tidal volumes – mL 500.0 (454.2 – 550.5) 500.0 (450.0 – 550.0) 527.7 (487.1 –

600.0) < 0.0001 500.0 (452.0 – 550.0)

500.0 (460.0 – 552.7) 0.247 500.0 (452.5 –

550.0)500.0 (456.0 – 559.9) 0.155

Tidal volumes – mL/kg PBW 8.1 (7.2 – 9.1) 8.0 (7.2 – 9.0) 9.2 (8.0 – 10.5) < 0.0001 8.1 (7.2 – 9.0) 8.4 (7.5 – 9.4) < 0.0001 8.1 (7.2 – 9.1) 8.2 (7.4 – 9.2) 0.0001

Tidal volumes – mL/kg ABW 6.7 (5.8 – 7.6) 6.8 (6.0 – 7.7) 5.0 (4.4 – 5.6) < 0.0001 6.7 (5.9 – 7.6) 6.7 (5.7 – 7.6) 0.110 6.7 (5.8 – 7.7) 6.7 (5.8 – 7.5) 0.171

PEEP – cm H2O 4.0 (0.0 – 5.0) 4.0 (0.0 – 5.0) 5.0 (2.0 – 5.0) < 0.0001 3.5 (0.0 – 5.0) 4.0 (1.5 – 5.0) < 0.0001 3.0 (0.0 – 5.0) 5.0 (2.0 – 5.0) < 0.0001

Respiratory rate – breaths/min 12.0 (12.0 – 13.0) 12.0 (12.0 – 13.0) 12.0 (12.0 –

14.0) < 0.0001 12.0 (11.5 – 13.0) 12.0 (12.0 – 14.0) < 0.0001 12.0 (12.0 – 13.0) 12.0 (12.0 – 13.0) 0.027

Minute ventilation – mL/min 6049 (5400 – 6960) 6000 (5400 – 6881) 6750 (6000 –

7560) < 0.0001 6000 (5400 – 6864) 6453 (5722 – 7200) < 0.0001 6000 (5400 – 6900) 6160 (5500 – 7020) 0.002

Ppeak – cm H2O 17.5 (15.0 – 21.0) 17.0 (15.0 – 20.0) 23.0 (20.0 – 26.0) < 0.0001 17.0 (14.5 – 20.0) 19.5 (16.5 – 23.5) < 0.0001 17.0 (14.5 – 20.0) 19.0 (16.0 – 22.0) < 0.0001

Cdyn.calc. – mL/cm H2O 35.3 (28.6 – 43.0) 35.7 (2.93 – 43.6) 28.9 (23.7 –

35.1) < 0.0001 36.1 (29.4 – 44.1) 31.2 (25.5 – 38.4) < 0.0001 35.7 (29.1 – 43.7) 33.8 (27.3 – 41.2) < 0.0001

Cq.stat. – mL/cm H2O 41.8 (34.0 – 51.5) 42.8 (34.8 – 52.3) 33.9 (28.2 –

42.0) < 0.0001 43.3 (35.3 – 52.5) 36.5 (29.5 – 46.1) < 0.0001 42.6 (34.6 – 52.2) 40.0 (32.3 – 50.0) < 0.0001

Recruitment manoeuvre performed 9.5 (779/82141) 8.7 (653/7507) 17.6 (126/716) < 0.0001 8.3 (550/6636) 14.3 (229/1605) < 0.0001 7.9 (484/6097) 13.8 (295/2144) < 0.0001

FiO2 0.51 (0.45 – 0.70) 0.50 (0.45 – 0.70) 0.55 (0.50 – 0.72) < 0.0001 0.50 (0.45 – 0.70) 0.54 (0.48 – 0.70) 0.002 0.54 (0.46 – 0.72) 0.50 (0.45 – 0.60) < 0.0001

< 0.40 7.0 (577/8241) 7.3 (549/7507) 3.8 (27/716)

< 0.0001

7.3 (484/6636) 5.8 (93/1605)

0.014

6.5 (398/6097) 8.3 (179/2144)

< 0.0001>= 0.40 – < 0.60 52.9 (4358/8241) 53.0 (3977/7507) 52.0 (372/716) 53.2 (3531/6636) 51.5 (827/1605) 49.5 (3017/6097) 62.5 (1341/2144)>= 0.60 – < 0.80 29.9 (2460/8241) 29.8 (2238/7507) 30.2 (216/716) 29.6 (1966/6636) 30.8 (494/1605) 32.4 (1974/6097) 22.7 (486/2144)≥ 0.80 10.3 (846/8241) 9.9 (743/7507) 14.1 (101/716) 9.9 (655/6636) 11.9 (191/1605) 11.6 (708/6097) 6.4 (138/2144)

SpO2 – % 99.0 (98.0 – 100.0) 99.0 (98.0 – 100.0) 98.0 (97.0 – 99.0) < 0.0001 99.0 (98.0 – 100.0) 99.0 (98.0 – 100.0) < 0.0001 99.0 (98.0 – 100.0) 99.0 (98.0 –

100.0) 0.122

≥ 96 97.5 (8036/8240) 98.0 (7353/7506) 93.2 (667/716)< 0.0001

97.5 (6472/6635) 97.4 (1564/1605)0.765

97.8 (5961/6097) 96.8 (2075/2143)0.016> 90 – < 96 2.4 (195/8240) 1.9 (145/7506) 6.7 (48/716) 2.3 (155/6635) 2.5 (40/1605) 2.1 (128/6097) 3.1 (67/2143)

≤ 90 0.1 (9/8240) 0.1 (8/7506) 0.1 (1/716) 0.1 (8/6635) 0.1 (1/1605) 0.1 (8/6097) 0.0 (1/2143)

etCO2 – mmHg 33.7 (31.0 – 36.5) 33.7 (31.0 – 36.4) 35.0 (32.0 – 38.0) < 0.0001 33.7 (31.0 – 36.0) 34.5 (32.0 – 37.5) < 0.0001 34.0 (31.0 – 36.7) 33.4 (30.5 – 36.0) < 0.0001

Data is presented as median (QR) or proportion % (n/N); *Chi-square for categorical variables and Mann-Whitney for continuous variables; $Other (e.g. high frequency oscillatory ventilation, jet ventilation, synchronized intermittent mandatory ventilation (SIMV); PBW: predicted body weight, calculated as: 50 + 0.91 x (centimetres of height – 152.4) for males and 45.5 + 0.91 x (centimetres of height – 152.4) for females; ABW: actual body weight; PEEP: positive end-expiratory pressure; Cdyn.calc.: calculated dynamic respiratory compliance: Cdyn = tidal volume / (peak pressure minus PEEP); Cq.stat.: static respiratory compliance; FiO2: fraction inspired oxygen; SpO2: peripheral oxygen saturation; etCO2: expiratory carbon dioxide

LAS VEGAS study

Chap

ter

5

97

Table 2. Intraoperative ventilation characteristics

Variable All patients BMI < 35 BMI ≥ 35 p value* Non-laparoscopic Laparoscopic p value* ARISCAT < 26 ARISCAT ≥ 26 p value*

Ventilation mode

Volume Control 69.8 (5683/8147) 70.1 (5204/7421) 65.9 (467/709)

0.041

69.0 (4525/6558) 72.9 (1158/1589)

0.005

68.9 (4154/6026) 72.1 (1529/2121)

< 0.0001Pressure Control 17.0 (1383/8147) 16.8 (1250/7421) 18.3 (130/709) 17.3 (1132/6558) 15.8 (251/1589) 17.9 (1081/6026) 14.2 (302/2121)Pressure Support or Spontaneous 5.9 (482/8147) 5.8 (431/7421) 7.1 (50/709) 6.3 (412/6558) 4.4 (70/1589) 6.2 (373/6026) 5.2 (109/2121)

Other$ 7.4 (599/8147) 7.2 (536/7421) 8.7 (62/709) 7.5 (489/6558) 6.9 (110/1589) 6.9 (418/6026) 8.5 (181/2121)Tidal volumes – mL 500.0 (454.2 – 550.5) 500.0 (450.0 – 550.0) 527.7 (487.1 –

600.0) < 0.0001 500.0 (452.0 – 550.0)

500.0 (460.0 – 552.7) 0.247 500.0 (452.5 –

550.0)500.0 (456.0 – 559.9) 0.155

Tidal volumes – mL/kg PBW 8.1 (7.2 – 9.1) 8.0 (7.2 – 9.0) 9.2 (8.0 – 10.5) < 0.0001 8.1 (7.2 – 9.0) 8.4 (7.5 – 9.4) < 0.0001 8.1 (7.2 – 9.1) 8.2 (7.4 – 9.2) 0.0001

Tidal volumes – mL/kg ABW 6.7 (5.8 – 7.6) 6.8 (6.0 – 7.7) 5.0 (4.4 – 5.6) < 0.0001 6.7 (5.9 – 7.6) 6.7 (5.7 – 7.6) 0.110 6.7 (5.8 – 7.7) 6.7 (5.8 – 7.5) 0.171

PEEP – cm H2O 4.0 (0.0 – 5.0) 4.0 (0.0 – 5.0) 5.0 (2.0 – 5.0) < 0.0001 3.5 (0.0 – 5.0) 4.0 (1.5 – 5.0) < 0.0001 3.0 (0.0 – 5.0) 5.0 (2.0 – 5.0) < 0.0001

Respiratory rate – breaths/min 12.0 (12.0 – 13.0) 12.0 (12.0 – 13.0) 12.0 (12.0 –

14.0) < 0.0001 12.0 (11.5 – 13.0) 12.0 (12.0 – 14.0) < 0.0001 12.0 (12.0 – 13.0) 12.0 (12.0 – 13.0) 0.027

Minute ventilation – mL/min 6049 (5400 – 6960) 6000 (5400 – 6881) 6750 (6000 –

7560) < 0.0001 6000 (5400 – 6864) 6453 (5722 – 7200) < 0.0001 6000 (5400 – 6900) 6160 (5500 – 7020) 0.002

Ppeak – cm H2O 17.5 (15.0 – 21.0) 17.0 (15.0 – 20.0) 23.0 (20.0 – 26.0) < 0.0001 17.0 (14.5 – 20.0) 19.5 (16.5 – 23.5) < 0.0001 17.0 (14.5 – 20.0) 19.0 (16.0 – 22.0) < 0.0001

Cdyn.calc. – mL/cm H2O 35.3 (28.6 – 43.0) 35.7 (2.93 – 43.6) 28.9 (23.7 –

35.1) < 0.0001 36.1 (29.4 – 44.1) 31.2 (25.5 – 38.4) < 0.0001 35.7 (29.1 – 43.7) 33.8 (27.3 – 41.2) < 0.0001

Cq.stat. – mL/cm H2O 41.8 (34.0 – 51.5) 42.8 (34.8 – 52.3) 33.9 (28.2 –

42.0) < 0.0001 43.3 (35.3 – 52.5) 36.5 (29.5 – 46.1) < 0.0001 42.6 (34.6 – 52.2) 40.0 (32.3 – 50.0) < 0.0001

Recruitment manoeuvre performed 9.5 (779/82141) 8.7 (653/7507) 17.6 (126/716) < 0.0001 8.3 (550/6636) 14.3 (229/1605) < 0.0001 7.9 (484/6097) 13.8 (295/2144) < 0.0001

FiO2 0.51 (0.45 – 0.70) 0.50 (0.45 – 0.70) 0.55 (0.50 – 0.72) < 0.0001 0.50 (0.45 – 0.70) 0.54 (0.48 – 0.70) 0.002 0.54 (0.46 – 0.72) 0.50 (0.45 – 0.60) < 0.0001

< 0.40 7.0 (577/8241) 7.3 (549/7507) 3.8 (27/716)

< 0.0001

7.3 (484/6636) 5.8 (93/1605)

0.014

6.5 (398/6097) 8.3 (179/2144)

< 0.0001>= 0.40 – < 0.60 52.9 (4358/8241) 53.0 (3977/7507) 52.0 (372/716) 53.2 (3531/6636) 51.5 (827/1605) 49.5 (3017/6097) 62.5 (1341/2144)>= 0.60 – < 0.80 29.9 (2460/8241) 29.8 (2238/7507) 30.2 (216/716) 29.6 (1966/6636) 30.8 (494/1605) 32.4 (1974/6097) 22.7 (486/2144)≥ 0.80 10.3 (846/8241) 9.9 (743/7507) 14.1 (101/716) 9.9 (655/6636) 11.9 (191/1605) 11.6 (708/6097) 6.4 (138/2144)

SpO2 – % 99.0 (98.0 – 100.0) 99.0 (98.0 – 100.0) 98.0 (97.0 – 99.0) < 0.0001 99.0 (98.0 – 100.0) 99.0 (98.0 – 100.0) < 0.0001 99.0 (98.0 – 100.0) 99.0 (98.0 –

100.0) 0.122

≥ 96 97.5 (8036/8240) 98.0 (7353/7506) 93.2 (667/716)< 0.0001

97.5 (6472/6635) 97.4 (1564/1605)0.765

97.8 (5961/6097) 96.8 (2075/2143)0.016> 90 – < 96 2.4 (195/8240) 1.9 (145/7506) 6.7 (48/716) 2.3 (155/6635) 2.5 (40/1605) 2.1 (128/6097) 3.1 (67/2143)

≤ 90 0.1 (9/8240) 0.1 (8/7506) 0.1 (1/716) 0.1 (8/6635) 0.1 (1/1605) 0.1 (8/6097) 0.0 (1/2143)

etCO2 – mmHg 33.7 (31.0 – 36.5) 33.7 (31.0 – 36.4) 35.0 (32.0 – 38.0) < 0.0001 33.7 (31.0 – 36.0) 34.5 (32.0 – 37.5) < 0.0001 34.0 (31.0 – 36.7) 33.4 (30.5 – 36.0) < 0.0001

Data is presented as median (QR) or proportion % (n/N); *Chi-square for categorical variables and Mann-Whitney for continuous variables; $Other (e.g. high frequency oscillatory ventilation, jet ventilation, synchronized intermittent mandatory ventilation (SIMV); PBW: predicted body weight, calculated as: 50 + 0.91 x (centimetres of height – 152.4) for males and 45.5 + 0.91 x (centimetres of height – 152.4) for females; ABW: actual body weight; PEEP: positive end-expiratory pressure; Cdyn.calc.: calculated dynamic respiratory compliance: Cdyn = tidal volume / (peak pressure minus PEEP); Cq.stat.: static respiratory compliance; FiO2: fraction inspired oxygen; SpO2: peripheral oxygen saturation; etCO2: expiratory carbon dioxide

98

Table 3. Intraoperative complications

Variable All patients BMI < 35 BMI ≥ 35 p value* Non laparoscopic Laparoscopic p-value* ARISCAT < 26 ARISCAT ≥ 26 p value*

Any de–saturation 4.0 (331/8227) 3.4 (252/7496) 10.8 (77/714) < 0.0001 4.1 (274/6627) 3.6 (57/1600) 0.295 3.3 (202/6092) 6.0 (129/2135) < 0.0001

Unplanned recruitment manoeuvre

3.4 (282/8222) 3.0 (225/7492) 7.9 (56/713) < 0.0001 3.2 (214/6624) 4.3 (68/1598) 0.043 2.5 (151/6087) 6.1 (131/2135) < 0.0001

Ventilatory pressure reduction 2.8 (234/8216) 2.3 (172/7485) 8.4 (60/714) < 0.0001 2.0

(132/6621) 6.4 (102/1595) < 0.0001 2.1 (127/6087) 5.0 (107/2129) < 0.0001

Expiratory flow limitation 0.5 (43/8180) 0.5 (34/7456) 1.3 (9/707) 0.004 0.4 (27/6590) 1.0 (16/1590) 0.003 0.4

(23/6063) 0.9 (20/2117) 0.0001

Hypotension 27.4 (2251/8229) 27.3 (2046/7498) 27.9 (199/714) 0.738 29.1 (1929/6629) 20.1 (322/1600) < 0.0001 23.5 (1433/6092) 38.3 (818/2137) < 0.0001

Vaso-active drugs 23.6 (1938/8229) 23.4 (1754/7498) 24.9 (178/714) 0.354 25.1 (1664/6629) 17.1 (274/1600) < 0.0001 18.7 (1139/6092) 37.4 (799/2137) < 0.0001

New arrhythmias 0.6 (48/8223) 0.6 (44/7492) 0.4 (3/714) 0.571 0.6 (41/6625) 0.4 (7/1598) 0.394 0.3 (18/6088) 1.4 (30/2135) < 0.0001

All complications presented as proportion % (n/N); *Chi-square for categorical variables; Definitions intraoperative complications: Any de–saturation: defined as occurrence of SpO2 < 92%; Unplanned recruitment manoeuvre: ventilation strategies aimed to restore lung aeration; ventilation pressure reduction: ventilation strategies aimed to lower peak and/or plateau pressures; Expiratory flow limitation: defined as expiratory flow higher than zero at end-expiration as suggested by visual analysis of the flow curve; Hypotension: defined as SAP < 90mmHg for 3 min or longer; Need for vaso-active drugs: any vaso-active drug given to correct hypotension; New onset arrhythmias: defined as new onset of atrial fibrillation, sustained ventricular tachycardia, supraventricular tachycardia, and ventricular fibrillation

Table 4. Postoperative outcome measures

Variable All patients BMI < 35 BMI ≥ 35 p value* Non-Laparoscopic Laparoscopic p value* ARISCAT < 26 ARISCAT ≥ 26 p value*

Total PPCs# 861 / 8241 (10.4) 756 / 7507 (10.1) 101 / 716 (14.1) < 0.0001 705 / 6636 (10.6) 156 / 1605 (9.7) 0.277 410 / 6097 (6.7) 451 / 2144 (21.0) < 0.0001

Need for O2 therapy$ 700 / 8241 (8.5) 611 / 7507 (8.1) 86 / 716 (12) < 0.0001 567 / 6636 (8.6) 133 / 1605 (8.3) 0.721 339 / 6097 (5.6) 361 / 2144 (16.8) < 0.0001

Respiratory failure 138 / 8241 (1.7) 113 / 7507 (1.5) 25 / 716 (3.5) < 0.0001 115 / 6636 (1.7) 23 / 1605 (1.4) 0.399 55 / 6097 (0.9) 83 / 2144 (3.9) < 0.0001

Mechanical ventilation 89 / 8241 (1.1) 80 / 7507 (1.1) 9 / 716 (1.3) 0.635 78 / 6636 (1.2) 11 / 1605 (0.7) 0.088 34 / 6097 (0.6) 55 / 2144 (2.6) < 0.0001

ARDS 9 / 8241 (0.1) 7 / 7507 (0.1) 2 / 716 (0.3) 0.150 9 / 6636 (0.1) 0 / 1605 (0) 0.139 1 / 6097 (0) 8 / 2144 (0.4) < 0.0001

Pneumonia 35 / 8241 (0.4) 29 / 7507 (0.4) 5 / 716 (0.7) 0.213 31 / 6636 (0.5) 4 / 1605 (0.2) 0.227 9 / 6097 (0.1) 26 / 2144 (1.2) < 0.0001

Pneumothorax 12 / 8241 (0.1) 12 / 7507 (0.2) 0 / 716 (0) 0.284 12 / 6636 (0.2) 0 / 1605 (0) 0.088 8 / 6097 (0.1) 4 / 2144 (0.2) 0.564

Hospital LOS 1 (0 – 4) 1 (0 – 4) 1 (0 – 4) 0.404 1 (0 – 4) 1 (0 – 3) < 0.0001 1 (0 – 3) 4 (1 – 5) < 0.0001

In-hospital mortality 42 / 7594 (0.5) 37 / 6917 (0.5) 4 / 661 (0.6) 0.813 40 / 6113 (0.7) 2 / 1481 (0.1) 0.015 5 / 5613 (0.1) 37 / 1981 (1.9) < 0.0001

Hospital-free days** 26 (23 – 27) 26 (23 – 27) 26 (23 – 27) 0.460 26 (23 – 27) 26 (24 – 27) < 0.0001 26 (24 – 27) 23 (21 – 26) < 0.0001

All data is presented as proportion, % (n/N) or median (QR); *Comparison of differences within a subgroup is performed by using the t-test for continuous variables; BMI: body mass index in kg/m2; ARISCAT: Assess Respiratory Risk in Surgical Patients in Catalonia (score); PPCs: Postoperative pulmonary complications; ARDS: the acute respiratory distress syndrome; LOS: length of hospital stay; PPCs: on day 1 to 5 were scored YES as soon as the event occurred on either ward or intensive care unit; #Total PPCs: one patient could present with multiple PPCs, but was scored only once (YES or NO principle); $Need for O2 therapy, when not given as standard of care; **Hospital-free days when discharged and alive at day 28

LAS VEGAS study

Chap

ter

5

99

Table 3. Intraoperative complications

Variable All patients BMI < 35 BMI ≥ 35 p value* Non laparoscopic Laparoscopic p-value* ARISCAT < 26 ARISCAT ≥ 26 p value*

Any de–saturation 4.0 (331/8227) 3.4 (252/7496) 10.8 (77/714) < 0.0001 4.1 (274/6627) 3.6 (57/1600) 0.295 3.3 (202/6092) 6.0 (129/2135) < 0.0001

Unplanned recruitment manoeuvre

3.4 (282/8222) 3.0 (225/7492) 7.9 (56/713) < 0.0001 3.2 (214/6624) 4.3 (68/1598) 0.043 2.5 (151/6087) 6.1 (131/2135) < 0.0001

Ventilatory pressure reduction 2.8 (234/8216) 2.3 (172/7485) 8.4 (60/714) < 0.0001 2.0

(132/6621) 6.4 (102/1595) < 0.0001 2.1 (127/6087) 5.0 (107/2129) < 0.0001

Expiratory flow limitation 0.5 (43/8180) 0.5 (34/7456) 1.3 (9/707) 0.004 0.4 (27/6590) 1.0 (16/1590) 0.003 0.4

(23/6063) 0.9 (20/2117) 0.0001

Hypotension 27.4 (2251/8229) 27.3 (2046/7498) 27.9 (199/714) 0.738 29.1 (1929/6629) 20.1 (322/1600) < 0.0001 23.5 (1433/6092) 38.3 (818/2137) < 0.0001

Vaso-active drugs 23.6 (1938/8229) 23.4 (1754/7498) 24.9 (178/714) 0.354 25.1 (1664/6629) 17.1 (274/1600) < 0.0001 18.7 (1139/6092) 37.4 (799/2137) < 0.0001

New arrhythmias 0.6 (48/8223) 0.6 (44/7492) 0.4 (3/714) 0.571 0.6 (41/6625) 0.4 (7/1598) 0.394 0.3 (18/6088) 1.4 (30/2135) < 0.0001

All complications presented as proportion % (n/N); *Chi-square for categorical variables; Definitions intraoperative complications: Any de–saturation: defined as occurrence of SpO2 < 92%; Unplanned recruitment manoeuvre: ventilation strategies aimed to restore lung aeration; ventilation pressure reduction: ventilation strategies aimed to lower peak and/or plateau pressures; Expiratory flow limitation: defined as expiratory flow higher than zero at end-expiration as suggested by visual analysis of the flow curve; Hypotension: defined as SAP < 90mmHg for 3 min or longer; Need for vaso-active drugs: any vaso-active drug given to correct hypotension; New onset arrhythmias: defined as new onset of atrial fibrillation, sustained ventricular tachycardia, supraventricular tachycardia, and ventricular fibrillation

Table 4. Postoperative outcome measures

Variable All patients BMI < 35 BMI ≥ 35 p value* Non-Laparoscopic Laparoscopic p value* ARISCAT < 26 ARISCAT ≥ 26 p value*

Total PPCs# 861 / 8241 (10.4) 756 / 7507 (10.1) 101 / 716 (14.1) < 0.0001 705 / 6636 (10.6) 156 / 1605 (9.7) 0.277 410 / 6097 (6.7) 451 / 2144 (21.0) < 0.0001

Need for O2 therapy$ 700 / 8241 (8.5) 611 / 7507 (8.1) 86 / 716 (12) < 0.0001 567 / 6636 (8.6) 133 / 1605 (8.3) 0.721 339 / 6097 (5.6) 361 / 2144 (16.8) < 0.0001

Respiratory failure 138 / 8241 (1.7) 113 / 7507 (1.5) 25 / 716 (3.5) < 0.0001 115 / 6636 (1.7) 23 / 1605 (1.4) 0.399 55 / 6097 (0.9) 83 / 2144 (3.9) < 0.0001

Mechanical ventilation 89 / 8241 (1.1) 80 / 7507 (1.1) 9 / 716 (1.3) 0.635 78 / 6636 (1.2) 11 / 1605 (0.7) 0.088 34 / 6097 (0.6) 55 / 2144 (2.6) < 0.0001

ARDS 9 / 8241 (0.1) 7 / 7507 (0.1) 2 / 716 (0.3) 0.150 9 / 6636 (0.1) 0 / 1605 (0) 0.139 1 / 6097 (0) 8 / 2144 (0.4) < 0.0001

Pneumonia 35 / 8241 (0.4) 29 / 7507 (0.4) 5 / 716 (0.7) 0.213 31 / 6636 (0.5) 4 / 1605 (0.2) 0.227 9 / 6097 (0.1) 26 / 2144 (1.2) < 0.0001

Pneumothorax 12 / 8241 (0.1) 12 / 7507 (0.2) 0 / 716 (0) 0.284 12 / 6636 (0.2) 0 / 1605 (0) 0.088 8 / 6097 (0.1) 4 / 2144 (0.2) 0.564

Hospital LOS 1 (0 – 4) 1 (0 – 4) 1 (0 – 4) 0.404 1 (0 – 4) 1 (0 – 3) < 0.0001 1 (0 – 3) 4 (1 – 5) < 0.0001

In-hospital mortality 42 / 7594 (0.5) 37 / 6917 (0.5) 4 / 661 (0.6) 0.813 40 / 6113 (0.7) 2 / 1481 (0.1) 0.015 5 / 5613 (0.1) 37 / 1981 (1.9) < 0.0001

Hospital-free days** 26 (23 – 27) 26 (23 – 27) 26 (23 – 27) 0.460 26 (23 – 27) 26 (24 – 27) < 0.0001 26 (24 – 27) 23 (21 – 26) < 0.0001

All data is presented as proportion, % (n/N) or median (QR); *Comparison of differences within a subgroup is performed by using the t-test for continuous variables; BMI: body mass index in kg/m2; ARISCAT: Assess Respiratory Risk in Surgical Patients in Catalonia (score); PPCs: Postoperative pulmonary complications; ARDS: the acute respiratory distress syndrome; LOS: length of hospital stay; PPCs: on day 1 to 5 were scored YES as soon as the event occurred on either ward or intensive care unit; #Total PPCs: one patient could present with multiple PPCs, but was scored only once (YES or NO principle); $Need for O2 therapy, when not given as standard of care; **Hospital-free days when discharged and alive at day 28

100

Second, in randomized controlled trials in abdominal surgery patients benefit was found from a bundle of interventions, namely higher levels of PEEP, recruitment manoeuvres and lower VT, making it impossible to determine which of these factors contributed to the protection against PPCs. Also, we cannot rule out the possibility that one of the elements of the bundle was not beneficial or even harmful, and that such effect might have been masked by the other elements. The results of our present analysis suggest that during low VT ventilation, a level of PEEP ≥ 7 cmH2O is associated with higher incidence of PPCs.

The association between increasing PEEP levels and the occurrence of PPCs suggest a possible detrimental effect of intraoperative PEEP to non–injured lungs, challenging a recent retrospective study.31 Several trials implied a possible benefit of higher levels of PEEP.8–10 Notably, these trials tested a combination of lower VT plus higher levels of PEEP, precluding any conclusion regarding what intervention effectively reduced PPCs. In fact, a level of PEEP of as high as 12 cm H2O did not protect against occurrence of PPCs in patients undergoing open abdominal surgery.32 Theoretically, too high levels of PEEP could increase the end–inspiratory pressures, and cause overdistention on lung tissue that remains aerated at the end of expiration. Indeed, in the present study increasing levels of peak pressure were associated with a higher incidence of PPCs.

Figure 3. The proportion of patients without development of any postoperative pulmonary complication by postoperative day 6 presented by a Kaplan-Meier graph for all patientsPatients with body mass index (BMI) lower than 35 kg/m2 versus equal or higher than 35 kg/m2 (P < 0.0001, log–rank test); patients undergoing laparoscopic versus non–laparoscopic surgery (P = 0.420, log–rank test); and patients with a low versus high risk for PPCs, according to the ARISCAT score (lower versus equal and higher than 26) (P < 0.0001, log–rank test)

LAS VEGAS study

Chap

ter

5

101Figure 4. PROBIT logistic regression analysisDose-response relationship curve between the level of PEEP (cm H2O) used during general anaesthesia for surgery and the probability of postoperative pulmonary complications. The line represents the cubic term fitting all the points

The observation that patients with a BMI ≥ 35 kg/m2 were ventilated with higher VT might be explained by the fact that patient height is not routinely measured during the preoperative visit, and easily overestimated in obese patients. The slightly higher level of PEEP and FiO2 levels used in patients with BMI ≥ 35 kg/m2, or undergoing laparoscopic surgery, likely reflect measures to prevent or treat atelectasis and hypoxemia. In contrast, the combination of higher levels of PEEP with lower FiO2 in patients with an ARISCAT score ≥ 26 might reflect the concept of protective ventilation in previously injured lungs.

This study knows several limitations. First, the observational character of this study does not make it possible to determine any cause-effect relationship. Second, willingness of participating centres to join the study may have caused a selection bias towards interest in protective ventilation, i.e., using low VT, during general anaesthesia for surgery. Third, any prospective observational study can interfere with daily practice, meaning that anaesthetists could have been more eager to use those ventilation settings that are considered lung–protective. Fourth, we had no restriction in number of centres per country, which could have caused an overrepresentation of countries. Fifth, the majority of the participating centres were teaching hospitals with a median of 600 hospital beds and 15 operating theatres. And last, we were not able to collect data on postoperative incidence of atelectasis.

Conclusions

The most common ventilator setting during non–thoracic surgery includes VT < 10 mL/kg and levels of PEEP of 0 or 5 cm H2O. Recruitment manoeuvres are seldom performed. The incidence of PPCs was as high as 10.4%, and independently associated with increasing PEEP levels, but not VT size.

102

Tabl

e 5.

Mul

tivar

iabl

e Lo

gisti

c Re

gres

sion

Usi

ng G

ener

aliz

ed L

inea

r Mix

ed M

odel

s

Varia

ble

Ove

rall

Coho

rtBM

I ≥ 3

5 kg

/m2

Lapa

rosc

opic

Sur

gery

ARIS

CAT

≥ 26

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Venti

latio

n se

tting

s

Tida

l vol

ume,

mL/

kg P

BW---

0.94

(0.8

1 –

1.08

), 0.

387

1.05

(0.9

1 –

1.22

), 0.

476

---

PEEP

, cm

H 2O

1.06

(1.0

1 –

1.12

), 0.

020

1.13

(1.0

1 –

1.26

), 0.

032

1.10

(0.9

9 –

1.22

), 0.

058

1.07

(1.0

1 –

1.14

), 0.

031

Peak

pre

ssur

e, c

mH 2

O1.

03 (1

.00

– 1.

06),

0.04

01.

09 (1

.03

– 1.

15),

0.00

31.

01 (0

.96

– 1.

07),

0.62

1---

F iO2

1.00

(0.9

9 –

1.01

), 0.

885

---1.

00 (0

.99

– 1.

01),

0.99

6---

Patie

nt c

hara

cter

istic

s

Mal

e se

x0.

99 (0

.79

– 1.

25),

0.96

6---

------

BMI,

kg/m

21.

01 (0

.99

– 1.

03),

0.08

1---

1.03

(0.9

9 –

1.06

), 0.

074

---

COPD

1.34

(0.9

7 –

1.86

), 0.

088

---0.

90 (0

.34

– 2.

38),

0.83

31.

68 (1

.06

– 2.

68),

0.02

8

Age,

yea

rs

≤ 50

51

– 8

0

> 80

1 (R

efer

ence

)1.

96 (1

.50

– 2.

59),

< 0.

0001

3.12

(1.7

9 –

5.42

), <

0.00

01

1 (R

efer

ence

)1.

89 (1

.03

– 3.

47),

0.03

92.

26 (0

.33

– 15

.59)

, 0.4

06

1 (R

efer

ence

)2.

00 (1

.24

– 3.

23),

0.00

41.

37 (0

.49

– 3.

82),

0.54

4

1 (R

efer

ence

)1.

70 (1

.17

– 2.

46),

0.00

52.

81 (1

.61

– 4.

88),

< 0.

0001

Func

tiona

l Sta

tus

In

depe

nden

t

Parti

ally

dep

ende

nt

Tota

lly d

epen

dent

1 (R

efer

ence

)1.

54 (1

.00

– 2.

35),

0.04

71.

93 (0

.95

– 3.

92),

0.06

9

---1

(Ref

eren

ce)

1.65

(0.5

8 –

4.70

), 0.

352

4.51

(0.3

2 –

63.4

8), 0

.264

1 (R

efer

ence

)1.

21 (0

.74

– 1.

98),

0.44

80.

99 (0

.32

– 3.

12),

0.99

7

Curr

ent s

mok

er0.

83 (0

.65

– 1.

05),

0.12

0---

------

Resp

irato

ry in

fecti

on1.

68 (1

.09

– 2.

58),

0.01

91.

84 (0

.77

– 4.

42),

0.17

02.

35 (0

.98

– 5.

62),

0.05

5---

Preo

pera

tive

SpO

2, %

96

91 –

95

90

1 (R

efer

ence

)1.

39 (1

.10

– 1.

76),

0.00

52.

55 (1

.26

– 5.

15),

0.00

9

1 (R

efer

ence

)1.

01 (0

.54

– 1.

89),

0.96

93.

09 (0

.86

– 11

.01)

, 0.0

82

1 (R

efer

ence

)1.

57 (0

.91

– 2.

69),

0.10

13.

81 (0

.47

– 30

.91)

, 0.2

10

1 (R

efer

ence

)1.

13 (0

.81

– 1.

57),

0.47

71.

83 (0

.93

– 3.

59),

0.07

9

Preo

pera

tive

anae

mia

1.92

(1.1

8 –

3.14

), 0.

009

---2.

58 (0

.76

– 8.

74),

0.12

7---

Surg

ical

cha

ract

eris

tics

Non

-lapa

rosc

opic

surg

ery

------

---0.

84 (0

.63

– 1.

14),

0.26

8

Urg

ency

of s

urge

ry

Elec

tive

U

rgen

cy

Emer

genc

y

1 (R

efer

ence

)1.

55 (1

.09

– 2.

21),

0.01

42.

63 (1

.44

– 4.

82),

0.00

2

1 (R

efer

ence

)1.

26 (0

.48

– 3.

33),

0.63

72.

21 (0

.37

– 13

.03)

, 0.3

79---

1 (R

efer

ence

)1.

76 (1

.07

– 2.

91),

0.02

72.

61 (1

.12

– 6.

08),

0.02

6

Plan

ned

dura

tion

of s

urge

ry, h

ours

2

2 –

3

> 3

1 (R

efer

ence

)1.

44 (1

.13

– 1.

83),

0.00

32.

16 (1

.60

– 2.

91),

< 0.

0001

1 (R

efer

ence

)1.

14 (0

.67

– 1.

93),

0.63

01.

31 (0

.55

– 3.

12),

0.53

5

1 (R

efer

ence

)1.

89 (0

.99

– 3.

62),

0.05

41.

98 (0

.91

– 4.

32),

0.08

6

1 (R

efer

ence

)1.

14 (0

.77

– 1.

69),

0.51

91.

31 (0

.78

– 2.

19),

0.30

7

Type

of i

ncis

ion

Pe

riphe

ral

Ab

dom

inal

1 (R

efer

ence

)1.

30 (0

.96

– 1.

75),

0.09

11

(Ref

eren

ce)

1.38

(0.7

9 –

2.41

), 0.

262

------

Intr

aope

rativ

e ch

arac

teris

tics

Flui

d in

take

, mill

ilitr

es

≤ 99

9

1000

– 1

999

20

00 –

299

9

≥ 30

00

1 (R

efer

ence

)1.

58 (1

.13

– 2.

23),

0.00

92.

19 (1

.41

– 3.

42),

0.00

12.

93 (1

.78

– 4.

82),

< 0.

0001

1 (R

efer

ence

)0.

99 (0

.47

– 2.

09),

0.99

21.

79 (0

.68

– 4.

74),

0.23

93.

19 (1

.10

– 9.

29),

0.03

3

1 (R

efer

ence

)1.

28 (0

.63

– 2.

62),

0.49

00.

93 (0

.28

– 3.

04),

0.90

72.

46 (0

.80

– 7.

57),

0.11

8

1 (R

efer

ence

)1.

62 (1

.00

– 2.

61),

0.04

91.

65 (0

.94

– 2.

90),

0.08

02.

21 (1

.24

– 3.

95),

0.00

7

Epid

ural

ana

esth

esia

1.22

(0.8

7 –

1.71

), 0.

253

1.84

(0.5

7 –

6.01

), 0.

309

2.56

(1.3

3 –

4.94

), 0.

005

1.02

(0.7

2 –

1.46

), 0.

894

No

use

of n

euro

mus

cula

r blo

ckin

g ag

ents

0.85

(0.5

8 –

1.23

), 0.

387

0.74

(0.2

2 –

2.54

), 0.

632

0.35

(0.0

3 –

4.00

), 0.

399

---

Desa

tura

tion

2.23

(1.4

1 –

3.54

), <

0.00

013.

34 (1

.59

– 7.

02),

0.00

12.

17 (0

.76

– 6.

16),

0.14

52.

20 (1

.27

– 3.

79),

0.00

5

LAS VEGAS study

Chap

ter

5

103

Tabl

e 5.

Mul

tivar

iabl

e Lo

gisti

c Re

gres

sion

Usi

ng G

ener

aliz

ed L

inea

r Mix

ed M

odel

s

Varia

ble

Ove

rall

Coho

rtBM

I ≥ 3

5 kg

/m2

Lapa

rosc

opic

Sur

gery

ARIS

CAT

≥ 26

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Venti

latio

n se

tting

s

Tida

l vol

ume,

mL/

kg P

BW---

0.94

(0.8

1 –

1.08

), 0.

387

1.05

(0.9

1 –

1.22

), 0.

476

---

PEEP

, cm

H 2O

1.06

(1.0

1 –

1.12

), 0.

020

1.13

(1.0

1 –

1.26

), 0.

032

1.10

(0.9

9 –

1.22

), 0.

058

1.07

(1.0

1 –

1.14

), 0.

031

Peak

pre

ssur

e, c

mH 2

O1.

03 (1

.00

– 1.

06),

0.04

01.

09 (1

.03

– 1.

15),

0.00

31.

01 (0

.96

– 1.

07),

0.62

1---

F iO2

1.00

(0.9

9 –

1.01

), 0.

885

---1.

00 (0

.99

– 1.

01),

0.99

6---

Patie

nt c

hara

cter

istic

s

Mal

e se

x0.

99 (0

.79

– 1.

25),

0.96

6---

------

BMI,

kg/m

21.

01 (0

.99

– 1.

03),

0.08

1---

1.03

(0.9

9 –

1.06

), 0.

074

---

COPD

1.34

(0.9

7 –

1.86

), 0.

088

---0.

90 (0

.34

– 2.

38),

0.83

31.

68 (1

.06

– 2.

68),

0.02

8

Age,

yea

rs

≤ 50

51

– 8

0

> 80

1 (R

efer

ence

)1.

96 (1

.50

– 2.

59),

< 0.

0001

3.12

(1.7

9 –

5.42

), <

0.00

01

1 (R

efer

ence

)1.

89 (1

.03

– 3.

47),

0.03

92.

26 (0

.33

– 15

.59)

, 0.4

06

1 (R

efer

ence

)2.

00 (1

.24

– 3.

23),

0.00

41.

37 (0

.49

– 3.

82),

0.54

4

1 (R

efer

ence

)1.

70 (1

.17

– 2.

46),

0.00

52.

81 (1

.61

– 4.

88),

< 0.

0001

Func

tiona

l Sta

tus

In

depe

nden

t

Parti

ally

dep

ende

nt

Tota

lly d

epen

dent

1 (R

efer

ence

)1.

54 (1

.00

– 2.

35),

0.04

71.

93 (0

.95

– 3.

92),

0.06

9

---1

(Ref

eren

ce)

1.65

(0.5

8 –

4.70

), 0.

352

4.51

(0.3

2 –

63.4

8), 0

.264

1 (R

efer

ence

)1.

21 (0

.74

– 1.

98),

0.44

80.

99 (0

.32

– 3.

12),

0.99

7

Curr

ent s

mok

er0.

83 (0

.65

– 1.

05),

0.12

0---

------

Resp

irato

ry in

fecti

on1.

68 (1

.09

– 2.

58),

0.01

91.

84 (0

.77

– 4.

42),

0.17

02.

35 (0

.98

– 5.

62),

0.05

5---

Preo

pera

tive

SpO

2, %

96

91 –

95

90

1 (R

efer

ence

)1.

39 (1

.10

– 1.

76),

0.00

52.

55 (1

.26

– 5.

15),

0.00

9

1 (R

efer

ence

)1.

01 (0

.54

– 1.

89),

0.96

93.

09 (0

.86

– 11

.01)

, 0.0

82

1 (R

efer

ence

)1.

57 (0

.91

– 2.

69),

0.10

13.

81 (0

.47

– 30

.91)

, 0.2

10

1 (R

efer

ence

)1.

13 (0

.81

– 1.

57),

0.47

71.

83 (0

.93

– 3.

59),

0.07

9

Preo

pera

tive

anae

mia

1.92

(1.1

8 –

3.14

), 0.

009

---2.

58 (0

.76

– 8.

74),

0.12

7---

Surg

ical

cha

ract

eris

tics

Non

-lapa

rosc

opic

surg

ery

------

---0.

84 (0

.63

– 1.

14),

0.26

8

Urg

ency

of s

urge

ry

Elec

tive

U

rgen

cy

Emer

genc

y

1 (R

efer

ence

)1.

55 (1

.09

– 2.

21),

0.01

42.

63 (1

.44

– 4.

82),

0.00

2

1 (R

efer

ence

)1.

26 (0

.48

– 3.

33),

0.63

72.

21 (0

.37

– 13

.03)

, 0.3

79---

1 (R

efer

ence

)1.

76 (1

.07

– 2.

91),

0.02

72.

61 (1

.12

– 6.

08),

0.02

6

Plan

ned

dura

tion

of s

urge

ry, h

ours

2

2 –

3

> 3

1 (R

efer

ence

)1.

44 (1

.13

– 1.

83),

0.00

32.

16 (1

.60

– 2.

91),

< 0.

0001

1 (R

efer

ence

)1.

14 (0

.67

– 1.

93),

0.63

01.

31 (0

.55

– 3.

12),

0.53

5

1 (R

efer

ence

)1.

89 (0

.99

– 3.

62),

0.05

41.

98 (0

.91

– 4.

32),

0.08

6

1 (R

efer

ence

)1.

14 (0

.77

– 1.

69),

0.51

91.

31 (0

.78

– 2.

19),

0.30

7

Type

of i

ncis

ion

Pe

riphe

ral

Ab

dom

inal

1 (R

efer

ence

)1.

30 (0

.96

– 1.

75),

0.09

11

(Ref

eren

ce)

1.38

(0.7

9 –

2.41

), 0.

262

------

Intr

aope

rativ

e ch

arac

teris

tics

Flui

d in

take

, mill

ilitr

es

≤ 99

9

1000

– 1

999

20

00 –

299

9

≥ 30

00

1 (R

efer

ence

)1.

58 (1

.13

– 2.

23),

0.00

92.

19 (1

.41

– 3.

42),

0.00

12.

93 (1

.78

– 4.

82),

< 0.

0001

1 (R

efer

ence

)0.

99 (0

.47

– 2.

09),

0.99

21.

79 (0

.68

– 4.

74),

0.23

93.

19 (1

.10

– 9.

29),

0.03

3

1 (R

efer

ence

)1.

28 (0

.63

– 2.

62),

0.49

00.

93 (0

.28

– 3.

04),

0.90

72.

46 (0

.80

– 7.

57),

0.11

8

1 (R

efer

ence

)1.

62 (1

.00

– 2.

61),

0.04

91.

65 (0

.94

– 2.

90),

0.08

02.

21 (1

.24

– 3.

95),

0.00

7

Epid

ural

ana

esth

esia

1.22

(0.8

7 –

1.71

), 0.

253

1.84

(0.5

7 –

6.01

), 0.

309

2.56

(1.3

3 –

4.94

), 0.

005

1.02

(0.7

2 –

1.46

), 0.

894

No

use

of n

euro

mus

cula

r blo

ckin

g ag

ents

0.85

(0.5

8 –

1.23

), 0.

387

0.74

(0.2

2 –

2.54

), 0.

632

0.35

(0.0

3 –

4.00

), 0.

399

---

Desa

tura

tion

2.23

(1.4

1 –

3.54

), <

0.00

013.

34 (1

.59

– 7.

02),

0.00

12.

17 (0

.76

– 6.

16),

0.14

52.

20 (1

.27

– 3.

79),

0.00

5

104

Varia

ble

Ove

rall

Coho

rtBM

I ≥ 3

5 kg

/m2

Lapa

rosc

opic

Sur

gery

ARIS

CAT

≥ 26

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Mul

tivar

iabl

e M

odel

OR

(95%

CI),

p v

alue

Arrh

ythm

ias

2.01

(0.7

8 –

5.22

), 0.

149

------

2.87

(1.0

5 –

7.86

), 0.

040

Hypo

tens

ion

1.04

(0.7

3 –

1.47

), 0.

833

1.54

(0.7

4 –

3.24

), 0.

250

0.96

(0.5

3 –

1.71

), 0.

882

1.20

(0.8

4 –

1.73

), 0.

319

Nee

d of

vas

oacti

ve d

rugs

1.08

(0.7

2 –

1.62

), 0.

691

0.62

(0.2

6 –

1.51

), 0.

291

0.84

(0.4

2 –

1.66

), 0.

608

0.97

(0.6

1 –

1.55

), 0.

903

Post

oper

ative

cha

ract

eris

tics

No

reve

rsal

of n

euro

mus

cula

r bl

ocka

de1.

32 (0

.99

– 1.

75),

0.05

4---

1.42

(0.8

5 –

2.38

), 0.

175

1.11

(0.7

7 –

1.62

), 0.

568

Post

oper

ative

resid

ual c

urar

isatio

n3.

39 (1

.57

– 7.

29),

0.00

24.

98 (0

.94

– 26

.46)

, 0.0

593.

30 (1

.20

– 9.

07),

0.02

13.

91 (1

.79

– 8.

55),

0.00

1

Varia

bles

with

a p

val

ue a

bove

0.2

in th

e un

ivar

iate

ana

lysis

wer

e ex

clud

ed fr

om th

e m

ultiv

aria

te a

naly

sis a

nd th

e ce

lls o

f the

se v

aria

bles

in th

is ta

ble

wer

e ke

pt b

lank

. CI,

confi

denc

e in

terv

al; B

MI:

body

mas

s in

dex;

ARS

ICAT

sco

re <

26

pred

icts

a lo

w ri

sk fo

r pos

tope

rativ

e pu

lmon

ary

com

plic

ation

s (P

PCs)

; ARI

SCAT

sco

re ≥

26

pred

icts

an

inte

rmed

iate

to h

igh

risk

for P

PCs;

PBW

, pre

dict

ed b

ody

wei

ght;

PEEP

, pos

itive

end

-exp

irato

ry p

ress

ure;

Pea

k pr

essu

re: p

eak

insp

irato

ry p

ress

ure;

FiO

2, in

spire

d fr

actio

n of

oxy

gen;

CO

PD: c

hron

ic o

bstr

uctiv

e pu

lmon

ary

dise

ase;

SpO

2: pe

riphe

ral o

xyge

n sa

tura

tion;

Hb:

hae

mog

lobi

n

LAS VEGAS study

Chap

ter

5

105

Table 6. Matched by propensity for PPC*

Variables Odds Ratio (95% CI) p value

Tidal volume, mL/kg PBW 0.96 (0.89 – 1.02) 0.178

PEEP, cmH2O 1.08 (1.04 – 1.13) < 0.0001

FiO2 0.99 (0.99 – 1.00) 0.599

Peak pressure, cmH2O 1.02 (1.00 – 1.04) 0.037

PPC, postoperative pulmonary complications; CI, confidence interval; PBW, predicted body weight; PEEP, positive end-expiratory pressure; FiO2, inspired fraction of oxygen; *Matched 1:3, PPC to No PPC, within propensity score of 0.15 or less. Matched by: age, sex, BMI, ARISCAT, preoperative SpO2, functional status, smoking, COPD, chronic co-morbidity, respiratory infection < 30 days, preoperative anaemia, type of surgery, planned duration of surgery, condition of surgery, type of incision, fluid intake, epidural anaesthesia, use of NMBA, reversal of NMBA and residual curarisation. Details on matching are found in eTable 9

106

References1. Mazo V, Sabate S, Canet J et al. Prospective external validation of a predictive score for postoperative pulmonary

complications. Anesthesiology 2014, 121(2):219-2312. Putensen C, Theuerkauf N, Zinserling J, Wrigge H, Pelosi P. Metaanalysis: ventilation strategies and outcomes of the

acute respiratory distress syndrome and acute lung injury. Ann Intern Med 2009, 151(8):566-5763. Serpa Neto A, Cardoso SO, Manetta JA et al. Association between use of lung-protective ventilation with lower tidal

volumes and clinical outcomes among patients without acute respiratory distress syndrome: a metaanalysis. JAMA 2012, 308(16):1651-1659

4. Serpa Neto A, Nagtzaam L, Schultz M. Ventilation with lower tidal volumes for critically ill patients without the acute respiratory distress syndrome: a systematic translational review and metaanalysis. Curr Opin Crit Care 2014, 20(1):25-32

5. Checkley W, Brower R, Korpak A, Thompson B. Effects of a clinical trial on mechanical ventilation practices in patients with acute lung injury. Am J Respir Crit Care Med 2008, 177(11):1215-1222

6. Esteban A, Frutos-Vivar F, Muriel A et al. Evolution of mortality over time in patients receiving mechanical ventilation. Am J Respir Crit Care Med 2013, 188(2):220-230

7. Needham DM, Colantuoni E, Mendez-Tellez PA et al. Lung protective mechanical ventilation and two year survival in patients with acute lung injury: prospective cohort study. BMJ 2012, 344:e2124

8. Severgnini P, Selmo G, Lanza C et al. Protective mechanical ventilation during general anesthesia for open abdominal surgery improves postoperative pulmonary function. Anesthesiology 2013, 118(6):1307-1321

9. Futier E, Constantin JM, Paugam-Burtz C et al. A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. N Engl J Med 2013, 369(5):428-437

10. Ge Y, Yuan L, Jiang X, Wang X, Xu R, Ma W. Effect of lung protection mechanical ventilation on respiratory function in the elderly undergoing spinal fusion. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2013, 38(1):81-85

11. Serpa Neto A, Barbas CS, Beiderlinden M, et al for the PROVE Network investigators. Protective ventilation with lower tidal volumes and high PEEP versus conventional ventilation with high tidal volume and low PEEP in patients under general anesthesia for surgery: A systematic review and individual patient data metaanalysis. Anesthesiology 2015, 123(1):66-78

12. Karalapillai D, Weinberg L, Galtieri J, et al. Current ventilation practice during general anaesthesia: a prospective audit in Melbourne, Australia. BMC Anesthesiol 2014;14:85

13. Wanderer JP, Ehrenfeld JM, Epstein RH, et al. Temporal trends and current practice patterns for intraoperative ventilation at U.S. academic medical centers: a retrospective study. BMC Anesthesiol 2015;15:40

14. Güldner A, Kiss T, Serpa Neto A, Hemmes SN et al, for the PROtective VEntilation Network. Intraoperative Protective Mechanical Ventilation for Prevention of Postoperative Pulmonary Complications - a Comprehensive Review of the Role of Tidal Volume, Positive End-Expiratory Pressure and Lung Recruitment Maneuvers. Anesthesiology 2015; 123(3): 692-713

15. International conference on harmonisation of technical requirements for registration of pharmaceuticals for human use: ICH Harmonised Tripartite Guideline: Guideline for Good Clinical Practice. J Postgrad Med 2001, 47(3):199-203

16. Arozullah AM, Daley J, Henderson WG, Khuri SF. Multifactorial risk index for predicting postoperative respiratory failure in men after major noncardiac surgery. The National Veterans Administration Surgical Quality Improvement Program. Ann Surg 2000, 232(2):242-253

17. Arozullah AM, Khuri SF, Henderson WG, Daley J. Development and validation of a multifactorial risk index for predicting postoperative pneumonia after major noncardiac surgery. Ann Intern Med 2001, 135(10):847-857

18. Smetana GW, Lawrence VA, Cornell JE. Preoperative pulmonary risk stratification for noncardiothoracic surgery: systematic review for the American College of Physicians. Ann Intern Med 2006, 144(8):581-595

19. Canet J, Gallart L, Gomar C et al. Prediction of Postoperative Pulmonary Complications in a Population-based Surgical Cohort. Anesthesiology 2010, 113(6):1338-1350

20. Bagley SC, White H, Golomb BA. Logistic regression in the medical literature: standards for use and reporting, with particular attention to one medical domain. J Clin Epidemiol 2001, 54(10):979-985

21. Hemmes SN, de Abreu MG, Pelosi P, Schultz MJ. ESA Clinical Trials Network 2012: LAS VEGAS - Local Assessment of Ventilatory Management during General Anaesthesia for Surgery and its effects on Postoperative Pulmonary Complications: a prospective, observational, international, multicentre cohort study. European journal of anaesthesiology 2013, 30(5):205-207

22. Austin PC. Balance diagnostics for comparing the distribution of baseline covariates between treatment groups in propensity-score matched samples. Stat Med 2009, 28(25):3083-3107

23. Kor DJ, Warner DO, Alsara A et al. Derivation and diagnostic accuracy of the surgical lung injury prediction model. Critical Care Medicine 2011, 115(1):117-128

24. Blum JM, Maile M, Park PK et al. A description of intraoperative ventilator management in patients with acute lung injury and the use of lung protective ventilation strategies. Anesthesiology 2010, 115(1):75-82

LAS VEGAS study

Chap

ter

5

107

25. Lellouche F, Dionne S, Simard S, Bussieres J, Dagenais F. High tidal volumes in mechanically ventilated patients increase organ dysfunction after cardiac surgery. Anesthesiology 2012, 116(5):1072-1082

26. Jaber S, Coisel Y, Chanques G et al. A multicentre observational study of intra-operative ventilatory management during general anaesthesia: tidal volumes and relation to body weight. Anaesthesia 2012, 67(9):999-1008

27. Talab HF, Zabani IA, Abdelrahman HS et al. Intraoperative ventilatory strategies for prevention of pulmonary atelectasis in obese patients undergoing laparoscopic bariatric surgery. Anesth Analg 2009, 109(5):1511-1516

28. Reinius H, Jonsson L, Gustafsson S et al. Prevention of atelectasis in morbidly obese patients during general anesthesia and paralysis: a computerized tomography study. Anesthesiology 2009, 111(5):979-987

29. Serpa Neto A, Simonis FD, Barbas CS et al. Association between tidal volume size, duration of ventilation, and sedation needs in patients without acute respiratory distress syndrome: an individual patient data metaanalysis. Intensive Care Medicine 2014, 40(7):950-957

30. Serpa Neto A, Hemmes SN, Barbas CS et al. Incidence of mortality and morbidity related to postoperative lung injury in patients who have undergone abdominal or thoracic surgery: a systematic review and metaanalysis. The Lancet Respiratory Medicine 2014, 2(12):1007-1015

31. Levin MA, McCormick PJ, Lin HM, Hosseinian L, Fischer GW. Low intraoperative tidal volume ventilation with minimal PEEP is associated with increased mortality. Br J Anaesth 2014, 113(1):97-108

32. PROVEnet, Hemmes SN, Gama de Abreu M, Pelosi P, Schultz MJ: High versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial. Lancet 2014, 384(9942):495-503

108

Supplementary Appendix to ‘Intraoperative Ventilation Strategies and Patient Outcomes Following Surgery: an International Observational Study (LAS VEGAS)’ List of PROVE Network InvestigatorsWriting committeeSabrine N.T. Hemmes (Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands); Ary Serpa Neto (Department of Critical Care Medicine; Hospital Israelita Albert Einstein; São Paulo, Brazil); Marcelo Gama de Abreu (University Hospital Dresden, Germany); Paolo Pelosi (IRCCS AOU San Martino IST Hospital, University of Genoa, Genoa, Italy); Marcus J. Schultz (Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands)

LAS VEGAS Steering Committee:Sabrine N.T. Hemmes (Academic Medical Center, Amsterdam, the Netherlands); Ary Serpa Neto (Department of Critical Care Medicine; Hospital Israelita Albert Einstein; São Paulo, Brazil); Jan M. Binnekade (Academic Medical Center, Amsterdam, the Netherlands); Jaume Canet (Hospital Universitari Germans Trias I Pujol, Barcelona, Spain); Goran Hedenstierna (University Hospital Uppsula, Uppsala, Sweden); Samir Jaber (Saint Eloi University Hospital, Montpellier, France); Michael Hiesmayr (Medical University Vienna, Vienna, Austria); Markus W. Hollmann (Academic Medical Center, Amsterdam, the Netherlands); Gary H. Mills (Sheffield Teaching Hospitals, Sheffield, U.K.); Marcos F. Vidal Melo (Massachusetts General Hospital, Boston, U.S.A.); Rupert Pearse (Queen Mary University of London, London, U.K.); Christian Putensen (University Hospital Bonn, Bonn, Germany); Werner Schmid (Medical University Vienna, Vienna, Austria); Paolo Severgnini (University of Insubria, Varese, Italy); Hermann Wrigge (University of Leipzig, Germany); Marcelo Gama de Abreu (University Hospital Dresden, Dresden, Germany); Paolo Pelosi (IRCCS AOU San Martino IST Hospital, University of Genoa, Genoa, Italy); Marcus J. Schultz (Academic Medical Center, Amsterdam, the Netherlands)

PROVE Network websitewww.provenet.eu

List of LAS VEGAS Network Collaborators

AustriaLKH Graz, Graz: Wolfgang Kroell, Helfried Metzler, Gerd Struber, Thomas WegscheiderAKH Linz, Linz: Hans GombotzMedical University Vienna: Michael Hiesmayr, Werner Schmid, Bernhard Urbanek

Belgium UCL - Cliniques Universitaires Saint Luc Brussels: David Kahn, Mona Momeni, Audrey Pospiech, Fernande Lois, Patrice Forget, Irina GrosuUniversitary Hospital Brussels (UZ Brussel): Jan Poelaert, Veerle van Mossevelde, Marie-Claire van MalderenHet Ziekenhuis Oost Limburg (ZOL), Genk: Dimitri Dylst, Jeroen van Melkebeek, Maud BeranGhent University Hospital, Gent: Stefan de Hert, Luc De Baerdemaeker, Bjorn Heyse, Jurgen Van Limmen, Piet Wyffels, Tom Jacobs, Nathalie Roels, Ann De BruyneMaria Middelares, Gent: Stijn van de Velde

Bosnia and Herzegovina General Hospital “prim Dr Abdulah Nakas” Sarajevo: Marina Juros-Zovko, Dejana Djonoviċ- Omanoviċ

CroatiaGeneral Hospital Cakovec, Cakovec: Selma PernarGeneral Hospital Karlovac, Karlovac: Josip Zunic, Petar Miskovic, Antonio Zilic University Clinical Hospital Osijek, Osijek: Slavica Kvolik, Dubravka Ivic, Darija Azenic-Venzera, Sonja Skiljic, Hrvoje Vinkovic, Ivana OputricUniversity Hospital Rijeka, Rijeka: Kazimir Juricic, Vedran FrkovicGeneral Hospital Dr J Bencevic, Slavonski Brod: Jasminka Kopic, Ivan Mirkovic

LAS VEGAS study

Chap

ter

5

109

University Hospital Center Split, Split: Nenad Karanovic, Mladen Carev, Natasa DropulicUniversity Hospital Merkur, Zagreb: Jadranka Pavicic Saric, Gorjana Erceg, Matea Bogdanovic DvorscakUniversity Hospital Sveti Duh, Zagreb: Branka Mazul-Sunko, Anna Marija Pavicic, Tanja GoranovicUniversity Hospital, Medical school, “Sestre milosrdnice” (Sister of Charity), Zagreb: Branka Maldini, Tomislav Radocaj, Zeljka Gavranovic, Inga Mladic-Batinica, Mirna Sehovic

Czech RepublicUniversity Hospital Brno, Brno: Petr Stourac, Hana Harazim, Olga Smekalova, Martina Kosinova, Tomas Kolacek, Kamil Hudacek, Michal Drab University Hospital Hradec Kralove, Hradec Kralove: Jan Brujevic, Katerina Vitkova, Katerina JirmanovaUniversity Hospital Ostrava, Ostrava: Ivana Volfova, Paula Dzurnakova, Katarina LiskovaNemocnice Znojmo, Znojmo: Radovan Dudas, Radek Filipsky

Egypt El Sahel Teaching hospital, Cairo: Samir el KafrawyKasr Al-Ainy Medical School, Cairo University: Hisham Hosny Abdelwahab, Tarek Metwally, Ahmed Abdel-RazekBeni Sueif University Hospital, Giza: Ahmed Mostafa El-Shaarawy, Wael Fathy Hasan, Ahmed Gouda AhmedFayoum University Hospital, Giza: Hany Yassin, Mohamed Magdy, Mahdy AbdelhadySuis medical Insurance Hospital, Suis: Mohamed Mahran

Estonia North Estonia Medical Center, Tallinn: Eiko Herodes, Peeter Kivik, Juri Oganjan, Annika AunTartu University Hospital, Tartu: Alar Sormus, Kaili Sarapuu, Merilin Mall, Juri Karjagin

France University Hospital of Clermont-Ferrand, Clermont-Ferrand: Emmanuel Futier, Antoine Petit, Adeline GerardInstitut Hospitalier Franco-Britannique, Levallois-Perret: Emmanuel Marret, Marc SolierSaint Eloi University Hospital, Montpellier: Samir Jaber, Albert Prades

Germany Fachkrankenhaus Coswig, Coswig: Jens Krassler, Simone MerzkyUniversity Hospital Carl Gustav Carus, Dresden: Marcel Gama de Abreu, Christopher Uhlig,Thomas Kiss, Anette Bundy, Thomas Bluth, Andreas Gueldner, Peter Spieth, Martin Scharffenberg, Denny Tran Thiem, Thea KochDuesseldorf University Hospital, Heinrich-Heine University: Tanja Treschan, Maximilian Schaefer, Bea Bastin, Johann Geib, Martin Weiss, Peter Kienbaum, Benedikt PannenDiakoniekrankenhaus Friederikenstift, Hannover: Andre Gottschalk, Mirja Konrad, Diana Westerheide, Ben SchwertfegerUniversity of Leipzig, Leipzig: Hermann Wrigge, Philipp Simon, Andreas Reske, Christian Nestler

Greece “Alexandra” general hospital of Athens, Athens: Dimitrios Valsamidis, Konstantinos Stroumpoulis General air force hospital, Athens: Georgios Anthopoulos, Antonis Andreaou, Dimitris Karapanos Aretaieion University Hospital, Athens: Kassiani Theodorak, Georgios Gkiokas, Marios-Konstantinos TtasoulisAttikon University Hospital, Athens: Tatiana Sidiropoulou, Foteini Zafeiropoulou, Panagiota Florou, Aggeliki PandaziAhepa University Hospital Thessaloniki, Thessaloniki: Georgia Tsaousig, Christos Nouris, Chryssa Pourzitaki,

Israel The Lady Davis Carmel Medical Center, Haifa: Dmitri Bystritski, Reuven Pizov, Arieh Eden

Italy Ospedale San. Paolo Bari, Bari: Caterina Valeria Pesce, Annamaria Campanile, Antonella MarrellaUniversity of Bari “Aldo Moro”, Bari: Salvatore Grasso, Michele De MicheleInstitute for Cancer Research and treatment, Candiolo, Turin: Francesco Bona, Gianmarco Giacoletto, Elena SardoAzienda Ospedaliera per l’emergenza Cannizzaro, Catania: Luigi Giancarlo Vicari SottosantiOspedale Melegnano, Cernuso, Milano: Maurizio SolcaAzienda Ospedaliera – Universitaria Sant’Anna, Ferrara: Carlo Alberto Volta, Savino Spadaro, Marco Verri, Riccardo

110

Ragazzi, Roberto ZoppellariOspedali Riuniti Di Foggia - University of Foggia, Foggia: Gilda Cinnella, Pasquale Raimondo, Daniela La Bella, Lucia Mirabella, Davide D’antiniIRCCS AOU San Martino IST Hospital, University of Genoa, Genoa: Paolo Pelosi, Alexandre Molin, Iole Brunetti, Angelo Gratarola, Giulia Pellerano, Rosanna Sileo, Stefano Pezzato, Luca MontagnaniIRCCS San Raffaele Scientific Institute, Milano: Laura Pasin, Giovanni Landoni, Alberto Zangrillo, Luigi Beretta, Ambra Licia Di Parma, Valentina Tarzia, Roberto Dossi, Marta Eugenia SassoneIstituto europeo di oncologia – ieo, Milano: Daniele Sances, Stefano Tredici, Gianluca Spano, Gianluca Castellani, Luigi Delunas, Sopio Peradze, Marco VenturinoOspedale Niguarda Ca’Granda Milano, Milano: Ines Arpino, Sara SherOspedale San Paolo - University of Milano, Milano: Concezione Tommasino, Francesca Rapido, Paola MorelliUniversity of Naples “Federico II” Naples: Maria Vargas, Giuseppe ServilloPoliclinico “P. Giaccone”, Palermo: Andrea Cortegiani, Santi Maurizio Raineri, Francesca Montalto, Vincenzo Russotto, Antonino GiarratanoAzienda Ospedaliero-Universitaria, Parma: Marco Baciarello, Michela Generali, Giorgia CeratiSanta Maria degli Angeli, Pordenone: Yigal LeykinOspedale Misericordia e Dolce - Usl4 Prato, Prato: Filippo Bressan, Vittoria Bartolini, Lucia ZamideiUniversity hospital of Sassari, Sassari: Luca Brazzi, Corrado Liperi, Gabriele Sales, Laura PistiddaInsubria University, Varese: Paolo Severgnini, Elisa Brugnoni, Giuseppe Musella, Alessandro Bacuzzi

Republic of Kosovo Distric hospital Gjakova, Gjakove: Dalip MuhardriUniversity Clinical Center of Kosova, Prishtina: Agreta Gecaj-Gashi, Fatos SadaRegional Hospital ”Prim.Dr. Daut Mustafa”, Prizren: Adem Bytyqi

Lithuania Medical University Hospital, Hospital of Lithuanian University of Health Sciences, Kaunas: Aurika Karbonskiene, Ruta Aukstakalniene, Zivile Teberaite, Erika SalciuteVilnius University Hospital - Institute of Oncology, Vilnius: Renatas Tikuisis, Povilas MiliauskasVilnius University Hospital - Santariskiu Clinics, Vilnius: Sipylaite Jurate, Egle Kontrimaviciute, Gabija TomkuteMalta Mater Dei Hospital, Msida: John Xuereb, Maureen Bezzina, Francis Joseph Borg

Netherlands Academic Medical Centre, University of Amsterdam: Sabrine Hemmes, Marcus Schultz, Markus Hollmann, Irene Wiersma, Jan Binnekade, Lieuwe BosVU University Medical Center, Amsterdam: Christa Boer, Anne DuvekotMC Haaglanden, Den Haag: Bas in ‘t Veld, Alice Werger, Paul Dennesen, Charlotte SeverijnsWestfriesgasthuis, Hoorn: Jasper De Jong, Jens Hering, Rienk van Beek

NorwayHaukeland University Hospital, Bergen: Stefan Ivars, Ib JammerFørde Central Hospital /Førde Sentral Sykehus, Førde: Alena BreidablikMartina Hansens Hospital, Gjettum: Katharina Skirstad Hodt, Frode Fjellanger, Manuel Vico AvalosBærum Hospital, Vestre Viken, Rud: Jannicke Mellin-Olsen, Elisabeth AnderssonStavanger University Hospital, Stavanger: Amir Shafi-Kabiri

PanamaHospital Santo Tomás, Panama: Ruby Molina, Stanley Wutai, Erick Morais

PortugalHospital do Espírito Santo - Évora, E.P.E, Évora.: Gloria Tareco, Daniel Ferreira, Joana AmaralCentro Hospitalar de Lisboa Central, E.P.E, Lisboa.: Maria de Lurdes Goncalves Castro, Susana Cadilha, Sofia AppletonCentro Hospitalar de Lisboa Ocidental, E.P.E. Hospital de S. Francisco Xavier, Lisboa: Suzana Parente, Mariana Correia, Diogo MartinsSantarem Hospital, Santarem: Angela Monteirosa, Ana Ricardo, Sara Rodrigues

LAS VEGAS study

Chap

ter

5

111

RomaniaSpital Orasenesc, Bolintin Vale: Lucian HorhotaClinical Emergency Hospital of Bucharest, Bucharest: Ioana Marina Grintescu, Liliana Mirea, Ioana Cristina GrintescuElias University Emergency Hospital, Bucharest: Dan Corneci, Silvius Negoita, Madalina Dutu, Ioana Popescu GarotescuEmergency Institute of Cardiovascular Diseases Inst. ‘’Prof. C. C. Iliescu’’, Bucharest: Daniela Filipescu, Alexandru Bogdan ProdanFundeni Clinical institute - Anaesthesia and Intensive Care, Bucharest: Gabriela Droc, Ruxandra Fota, Mihai PopescuFundeni Clinical institute - Intensive Care Unit, Bucharest: Dana Tomescu, Ana Maria Petcu, Marian Irinel TudoroiuHospital Profesor D Gerota, Bucharest: Alida Moise, Catalin-Traian GuranConstanta County Emergency Hospital, Constanta: Iorel Gherghina, Dan Costea, Iulia CindeaUniversity Emergency County Hospital Targu Mures, Targu Mures: Sanda-Maria Copotoiu, Ruxandra Copotoiu, Victoria Barsan, Zsolt Tolcser, Magda Riciu, Gheorghe Moldovan Septimiu, Mihaly Veres

Russia Krasnoyarsk State Medical University, Krasnoyarsk: Alexey Gritsan, Tatyana Kapkan, Galina Gritsan, Oleg KorolkovBurdenko Neurosurgery Institute, Moscow: Alexander Kulikov, Andrey LubninMoscow Regional Research Clinical Institute, Moscow: Alexey Ovezov, Pavel Prokoshev, Alexander Lugovoy, Natalia AnipchenkoMunicipal Clinical Hospital 7, Moscow: Andrey Babayants, Irina Komissarova, Karginova ZalinaReanimatology Research Institute n.a. Negovskij RAMS, Moscow: Valery Likhvantsev, Sergei Fedorov

Serbia Clinical Center of Vojvodina, Emergency Center, Novisad: Aleksandra Lazukic, Jasmina Pejakovic, Dunja Mihajlovic

SlovakiaNational Cancer Institute, Bratislava: Zuzana Kusnierikova, Maria ZelinkovaF.D. Roosevelt teaching Hospital, Banská Bystrica: Katarina Bruncakova, Lenka PolakovicovaFaculty Hospital Nové Zámky, Nové Zámky: Villiam Sobona

SloveniaInstitute of Oncology Ljubljana, Ljubljana: Barbka Novak-Supe, Ana Pekle-Golez, Miroljub Jovanov, Branka StrazisarUniversity Medical Centre Ljubljana, Ljubljana: Jasmina Markovic-Bozic, Vesna Novak-Jankovic, Minca Voje, Andriy Grynyuk, Ivan Kostadinov, Alenka Spindler-Vesel

Spain Hospital Sant Pau, Barcelona: Victoria Moral, Mari Carmen Unzueta, Carlos Puigbo, Josep FavaHospital Universitari Germans Trias I Pujol, Barcelona: Jaume Canet, Enrique Moret, Monica Rodriguez Nunez, Mar Sendra, Andrea Brunelli, Frederic RodenasUniversity of Navarra, Pamplona: Pablo Monedero, Francisco Hidalgo Martinez, Maria Jose Yepes Temino, Antonio Martinez Simon, Ana de Abajo LarribaCorporacion Sanitaria Parc Tauli, Sabadell: Alberto Lisi, Gisela Perez, Raquel MartinezConsorcio Hospital General Universitario de Valencia, Valencia: Manuel Granell, Jose Tatay Vivo, Cristina Saiz Ruiz, Jose Antonio de Andres IbanezHospital Clinico Valencia, Valencia: Ernesto Pastor, Marina Soro, Carlos Ferrando, Mario Defez Hospital Universitario Rio Hortega, Valladolid: Cesar Aldecoa Alvares-Santullano, Rocio Perez, Jesus Rico

SwedenCentral Hospital in Kristianstad: Monir Jawad, Yousif Saeed, Lars Gillberg

TurkeyUfuk University Hospital Ankara, Ankara: Zuleyha Kazak Bengisun, Baturay Kansu KazbekAkdeniz University Hospital, Antalya: Nesil Coskunfirat, Neval Boztug, Suat Sanli, Murat Yilmaz, Necmiye HadimiogluIstanbul University, Istanbul medical faculty, Istanbul: Nuzhet Mert Senturk, Emre Camci, Semra Kucukgoncu, Zerrin Sungur, Nukhet SivrikozAcibadem University, Istanbul: Serpil Ustalar Ozgen, Fevzi ToramanMaltepe University, Istanbul: Onur Selvi, Ozgur Senturk, Mine Yildiz

112

Dokuz Eylül Universitesi Tip Fakültesi, Izmir: Bahar Kuvaki, Ferim Gunenc, Semih Kucukguclu, Sule OzbilginSifa University Hospital, İzmir: Jale Maral, Seyda CanliSelcuk University faculty of medicine, Konya: Oguzhan Arun, Ali Saltali, Eyup AydoganFatih Sultan Mehmet Eğitim Ve Araştirma Hastanesi, Istanbul: Fatma Nur Akgun, Ceren Sanlikarip, Fatma Mine Karaman

UkraineInstitute Of Surgery And Transplantology, Kiev: Andriy MazurZaporizhzhia State Medical University, Zaporizhzhia: Sergiy Vorotyntsev

United KingdomSWARM Research Collaborative: for full list of SWARM contributors please see www.ukswarm.comNorthern Devon Healthcare NHS Trust, Barnstaple: Guy Rousseau, Colin Barrett, Lucia StancombeGolden Jubilee National Hospital, Clydebank, Scotland: Ben Shelley, Helen ScholesDarlington Memorial Hospital, County Durham and Darlington Foundation NHS Trust, Darlington: James Limb, Amir Rafi, Lisa Wayman, Jill DeaneRoyal Derby Hospital, Derby: David Rogerson, John Williams, Susan Yates, Elaine RogersDorset County Hospital, Dorchester: Mark Pulletz, Sarah Moreton, Stephanie JonesThe Princess Alexandra NHS Hospital Trust, Essex: Suresh Venkatesh, Maudrian Burton, Lucy Brown, Cait GoodallRoyal Devon and Exeter NHS Foundation Trust, Exeter: Matthew Rucklidge, Debbie Fuller, Maria Nadolski, Sandeep KusreHospital James Paget University Hospital NHS Foundation Trust, Great Yarmouth: Michael Lundberg, Lynn Everett, Helen NuttRoyal Surrey County Hospital NHS Foundation Trust, Guildford: Maka Zuleika, Peter Carvalho, Deborah Clements, Ben Creagh-BrownKettering General Hospital NHS Foundation Trust, Kettering: Philip Watt, Parizade RaymodeBarts Health NHS Trust, Royal London Hospital, London: Rupert Pearse, Otto Mohr, Ashok Raj, Thais CrearyNewcastle Upon Tyne Hospitals NHS Trust The Freeman Hospital High Heaton, Newcastle upon Tyne: Ahmed Chishti, Andrea Bell, Charley Higham, Alistair Cain, Sarah Gibb, Stephen MowatDerriford Hospital Plymouth Hospitals NHS Trust, Plymouth: Danielle Franklin, Claire West, Gary Minto, Nicholas Boyd Royal Hallamshire Hospital, Sheffield: Gary Mills, Emily Calton, Rachel Walker, Felicity Mackenzie, Branwen Ellison, Helen RobertsMid Staffordshire NHS, Stafford: Moses Chikungwa, Clare JacksonMusgrove Park Hospital, Taunton: Andrew Donovan, Jayne Foot, Elizabeth HomanSouth Devon Healthcare NHS Foundation Trust /Torbay Hospital, Torquay, Torbay: Jane Montgomery, David Portch, Pauline Mercer, Janet Palme Royal Cornwall Hospital, Truro: Jonathan Paddle, Anna Fouracres, Amanda Datson, Alyson Andrew, Leanne WelchMid Yorkshire Hospitals NHS Trust; Pinderfields Hospital, Wakefield: Alastair Rose, Sandeep Varma, Karen Simeson Sandwell and West Birmingham NHS Trust, West Bromich: Mrutyunjaya Rambhatla, Jaysimha Susarla, Sudhakar Marri, Krishnan Kodaganallur, Ashok Das, Shivarajan Algarsamy, Julie ColleyYork Teaching Hospitals NHS Foundation Trust, York: Simon Davies, Margaret Szewczyk, Thomas Smith

United States University of Colorado School of Medicine/University of Colorado Hospital, Aurora: Ana Fernandez- Bustamante, Elizabeth Luzier, Angela AlmagroMassachusetts General Hospital, Boston: Marcos Vidal Melo, Luiz Fernando, Demet SulemanjiMayo Clinic, Rochester: Juraj Sprung, Toby Weingarten, Daryl Kor, Federica Scavonetto, Yeo Tze

AcknowledgementsWe are indebted to all participating research nurses, nurse anaesthetists, surgeons, other physicians and our patients. Without them LAS VEGAS would never have been successful.We also thank Brigitte Leva, Sandrine Damster, and Benoit Plichon from the Research Team at the European Society of Anaesthesiology for their expertise and professional help in coordinating the trial and cleaning the study data of LAS VEGAS.

FundingA research grant of the European Society of Anaesthesiology supported this trial.

LAS VEGAS study

Chap

ter

5

113

STROBE Statement checklist

114

Randomization ProcedureCentres with >180 eligible patients per week are offered the option to reduce their patient cohort by randomization. These centres are identified according to the number of eligible patients per week, as reported in the LAS VEGAS Site Survey.

Distribution:< 180 procedures per week: no randomization180 – 360 procedures per week: randomization to include 50% of their cohort> 360 procedures per week: randomization to include 25% of their cohortOnly eligible, planned patients are suitable for randomization. All Emergency procedures and all procedures performed outside office hours (evening/ night/weekend) must be included and therefore should not enter the randomization program.

Details on randomization:- Randomization program: ALEA (computerized)- Stratified per centre- 1 inlog provided per centre- Random blocks- Variables collected:*Centre name*Date of surgery*Urgency of procedure*Surgical Procedure (same categories as collected in CRF)*Planned duration of surgery (in hours)

LAS VEGAS study

Chap

ter

5

115

*Age patient (in years)*ASA score (1 to 5 or not available)- Output: INCLUDE vs EXCLUDE

How does it work in practice?1. Inform ESA ([email protected]) if you want to make use of the randomization program2. Make a list of all eligible patients (e.g. print-out of planned surgical procedures that day)3. Randomize each patient4. You will receive output: INCLUDE or EXCLUDE5. Make a list of included patients6. Excluded patients do not have to be recorded or filed7. ALEA patient number generated after randomization does not have to be recorded the Patient

Identification Number generated by OpenClinica should be used. This PIN (Study Subject ID) is produced and provided when an electronic CRF is created for the patient.

Multiple operations within 5 day follow-up:2 scenarios:1. If a patient is re-operated during hospital stay, the study is not stopped and follow-up continues

to discharge. This patient does not enter randomization again.2. If the patient is discharged from the hospital and is re-operated later (re-admission); follow-

up is finished the day of the first discharge. No randomization.

eFigure 1. Length of hospital admission (PPC versus no PPC) The proportion of patients alive and admitted to the hospital by postoperative day 28 compared between patients with any postoperative pulmonary complication versus patients without any postoperative pulmonary complication by postoperative day 5 as presented by a Kaplan-Meier graph (P < 0.0001, log–rank test)

116

eFigure 2. In-hospital survival (PPC versus No PPC)The proportion of patients without in-hospital mortality by postoperative day 28 compared between patients with any postoperative pulmonary complication versus patients without any postoperative pulmonary complication by postoperative day 5 as presented by a Kaplan-Meier graph (P < 0.0001, log-rank test)

eFigure 3. Tidal volumes for BMI (Actual versus Predicted Body Weight)Distribution of tidal volumes in mL/kg for actual body weight versus predicted body weight (PBW) in the different obesity classes according to BMI (kg/m2) presented as box plots (median, QR, and ranges). Underweight: BMI < 18.5 kg/m2; No obesity: BMI 18.5 – 24.9 kg/m2; Overweight: BMI 25.0 - 29.9 kg/m2; Obesity Class 1: BMI 30.0 to 34.9 kg/m2; Obesity Class 2: BMI 35.0 to 39.9 kg/m2; Morbid obesity: BMI ≥ 40 kg/m2

LAS VEGAS study

Chap

ter

5

117

eFigure 4. Probability of postoperative pulmonary complications according to peak pressurePROBIT logistic regression showing the dose-relationship curve between the peak pressure (cm H2O) used during general anaesthesia for surgery and the probability of postoperative pulmonary complications. The line represents the cubic term fitting all the points

eTable 1. Definitions of intraoperative complications

Any de–saturation

Defined as SpO2 < 92%

Need for unplanned recruitment manoeuvre

Defined as ventilation strategies aimed to restore aeration of the lungs

Need for ventilatory pressure reduction

Defined as ventilation strategies aimed to lower peak and plateau pressure

New onset of expiratory flow limitation

Defined as expiratory flow higher than zero at end-expiration as suggested by visual analysis of the flow curve

Hypotension

Defined as SAP < 90 mmHg for 3 min or longer

Need for vaso-active drugs

Any vaso-active drug given to correct hypotension as defined above

Any new arrhythmias

Defined as atrial fibrillation [AF], sustained ventricular tachycardia [VT], supraventricular tachycardia [SVT], or ventricular fibrillation [VF]*

*Atrial fibrillation is defined by characteristic absolute irregularity of R-R intervals and concurrent loss of identifiable P waves in the ECG recordings; Ventricular tachycardia is characterized by ≥3 consecutive QRS complexes with a wide QRS complex at a rate of >100 beats/min and duration of >30 seconds; Supraventricular tachycardia is identified as a narrow QRS complex (<0.12 seconds) and a rate of >180 beats/min; Ventricular fibrillation is defined as chaotic ventricular electrical discharge with marked variability in QRS cycle length, morphology, and amplitude

118

eTable 2. Definition recruitment manoeuvres

Incremental PEEP

Stepwise increase in PEEP at constant tidal volume, mostly in steps of 5 cmH20, until peak/plateau airway pressure of above 30 cmH20 is reached. PEEP is sustained for at least 3 breaths and then returned back to baseline ventilation

Tidal volume recruitment

Stepwise increase in tidal volume until peak/plateau airway pressure of above 30 cmH20 is reached at constant PEEP level. At least 3 breaths with the plateau pressure of above 30cmH20 are performed, before returning back to baseline ventilation

Combined tidal and PEEP recruitment

PEEP and tidal volume are both stepwise increased to reach a peak/plateau pressure above 30 cmH20. At least 3 breaths with the plateau pressure of above 30 cmH20 are performed, before returning back to baseline ventilation

Inspiratory holds

Also called CPAP manoeuvres. During this kind of manoeuvre a positive airway pressure above 30 cmH20 is applied for 10 to 30 seconds and then returned back to baseline ventilation

Sustained inflation with bag

Manual hyperinflation using balloon/bag

eTable 3. Definitions of postoperative pulmonary complications*

Unexpected need for oxygen therapy

Supplemental oxygen administered due to PaO2 < 60 mmHg or SpO2 < 90% in room air. This excludes oxygen supplementation given as standard care (e.g. directly after arrival in the PACU)

Respiratory failure

PaO2 < 60 mmHg or SpO2 < 90% despite oxygen therapy, or need for non-invasive mechanical ventilation (NIV)

Mechanical ventilation

Unplanned new or prolonged invasive or non–invasive mechanical ventilation, after discharge from the operation room

ARDS

Definition according to the Berlin definition of ARDS1

Pneumonia

Presence of a new or progressive radiographic infiltrate plus at least two of three clinical features; fever >38°C or >100.4 °F, leucocytosis or leukopenia (WBC count >12,000 cells/mm³ or <4,000 cells/mm³) and purulent secretions

Pneumothorax

Defined as air in the pleural space with no vascular bed surrounding the visceral pleura on the chest X–ray

*Adapted from Canet et al.2

LAS VEGAS study

Chap

ter

5

119

eTable 4. Full list of participating centres

Country City Institution # Patients

Austria Graz LKH Graz 51

Austria Linz AKH Linz 40

Austria Vienna Medical University Vienna 34

Belgium Brussels UCL - Cliniques Universitaires Saint Luc Brussels 119

Belgium Brussels Universitary Hospital Brussels (UZ Brussel) 27

Belgium Genk Het ZOL (Ziekenhuis Oost Limburg) 65

Belgium Gent Ghent University Hospital 154

Belgium Gent Maria Middelares Gent 10

Bosnia and Herzegovina Sarajevo General Hospital “prim Dr Abdulah Nakas” Sarajevo 48

Croatia Cakovec General Hospital Čakovec 40

Croatia Karlovac General Hospital Karlovac 28

Croatia Osijek University Clinical Hospital Osijek 125

Croatia RIJEKA University Hospital Rijeka 53

Croatia Slavonski Brod General Hospital Dr J Bencevic 46

Croatia Split University Hospital Centre Split 96

Croatia Zagreb University Hospital Merkur 48

Croatia Zagreb University Hospital Sveti Duh 59

Croatia Zagreb University Hospital, Medical school, “Sestre milosrdnice” (Sister of Charity) 85

Czech Republic Brno University Hospital Brno 149

Czech Republic Hradec Kralove University Hospital Hradec Kralove 82

Czech Republic Ostrava University Hospital Ostrava 102

Czech Republic Znojmo Nemocnice Znojmo, p.o. 62

Egypt Cairo El Sahel Teaching hospital 62

Egypt Cairo Kasr Al-Ainy Medical School, Cairo University 38

Egypt Giza Beni Sueif University Hospital 42

Egypt Giza Fayoum University Hospital 49

Egypt Suis Suis medical Insurance Hospital 48

Estonia Tallinn North Estonia Medical Centre 164

Estonia Tartu Tartu University Hospital 80

120

Country City Institution # Patients

France Clermont-Ferrand Estaing Hospital, University Hospital of Clermont-Ferrand 30

France Levallois-Perret Institut Hospitalier Franco-Britannique 21

France Montpellier Saint Eloi University Hospital, Montpellier 140

Germany Coswig Fachkrankenhaus Coswig Gmbh, centre for pneumology and thoracic surgery 10

Germany Dresden University Hospital Carl Gustav Carus 219

Germany Duesseldorf Duesseldorf University Hospital, Heinrich-Heine University 129

Germany Hannover Diakoniekrankenhaus Friederikenstift 133

Germany Leipzig University of Leipzig 71

Greece Athens “Alexandra” general hospital of Athens 43

Greece Athens 251 General air force hospital 41

Greece Athens Aretaieion University Hospital 41

Greece Athens Attikon University Hospital 54

Greece Thessaloniki Ahepa University Hospital Thessaloniki 50

Israel Haifa The Lady Davis Carmel Medical Centre 40

Italy Bari Ospedale San. Paolo Bari 35

Italy Bari University of Bari “Aldo Moro” 81

Italy Candiolo (Turin) Institute for Cancer Research and treatment 28

Italy Catania Azienda Ospedaliera per l’emergenza Cannizzaro 39

Italy Cernuso (Milano) Ospedale Melegnano 5

Italy Ferrara Azienda Ospedaliera – Universitaria Sant’Anna (Ferrara) 88

Italy Foggia Ospedali Riuniti Di Foggia - University of Foggia 85

Italy Genoa IRCCS San Martino AOU IST Hospital, University of Genoa 173

Italy Milano IRCCS San Raffaele Scientific Institute 156

Italy Milano Istituto europeo di oncologia - ieo 131

Italy Milano Ospedale Niguarda Ca'Granda Milano 19

Italy Milano Ospedale San Paolo - University of Milano 46

Italy Naples University of Naples “Federico II” 57

Italy Palermo Policlinico “P. Giaccone” 92

LAS VEGAS study

Chap

ter

5

121

Country City Institution # Patients

Italy Parma Azienda Ospedaliero-Universitaria 81

Italy Pordenone Santa Maria degli Angeli 18

Italy Prato Ospedale Misericordia e Dolce - Usl4 Prato 41

Italy Sassari University hospital of Sassari 54

Italy Varese Insubria University 125

Lithuania Kaunas Kaunas Medical University Hospital, Hospital of Lithuanian University of Health Sciences 160

Lithuania Vilnius Vilnius University Hospital - Institute of Oncology 108

Lithuania Vilnius Vilnius University Hospital - Santariskiu Clinics 104

Malta Msida Mater Dei Hospital 27

Netherlands Amsterdam Academic Medical Centre, University of Amsterdam 183

Netherlands Amsterdam VU University Medical Centre 104

Netherlands Den Haag MC Haaglanden 94

Netherlands Hoorn Westfriesgasthuis 104

Norway Bergen Haukeland University Hospital 106

Norway Førde Førde Central Hospital /Førde Sentral Sykehus 58

Norway Gjettum Martina Hansens Hospital 57

Norway Rud Bærum Hospital, Vestre Viken 57

Norway Stavanger Stavanger University Hospital 70

Panama Panama Hospital Santo Tomás 40

Portugal Évora Hospital do Espírito Santo - Évora, EPE. 55

Portugal Lisboa Centro Hospitalar de Lisboa Central, EPE 49

Portugal Lisboa Centro Hospitalar de Lisboa Ocidental, EPE 41

Portugal Santarem Santarem Hospital 44

Republic of Kosovo Gjakovë Distric hospital Gjakova 9

Republic of Kosovo Prishtina University Clinical Centre of Kosova 92

Republic of Kosovo Prizren Regional Hospital ”Prim.Dr. Daut Mustafa” 39

Romania Bolintin Vale Spital orasenesc Bolintin Vale 8

Romania Bucharest Clinical Emergency Hospital of Bucharest 58

Romania Bucharest Elias University Emergency Hospital 53

122

Country City Institution # Patients

Romania Bucharest Emergency Institute of Cardiovascular Diseases Inst. ''Prof. C. C. Iliescu'' 6

Romania Bucharest Fundeni Clinical institute - Anaesthesia and Intensive Care 43

Romania Bucharest Fundeni Clinical institute - Intensive Care Unit 40

Romania Bucharest Hospital Profesor D Gerota 21

Romania Constanta Constanta County Emergency Hospital 73

Romania Targu Mures University Emergency County Hospital Targu Mures 138

Russia Krasnoyarsk Krasnoyarsk State Medical University 52

Russia Moscow Burdenko Neurosurgery Institute 60

Russia Moscow Moscow Regional Research Clinical Institute 59

Russia Moscow Municipal Clinical Hospital 7 49

Russia Moscow Reanimatology Research Institute n.a. Negovskij RAMS 61

Serbia Novisad Clinical Center of Vojvodina, Emergency Center 42

Slovakia Bratislava National Cancer Institute 18

Slovakia Banská Bystrica F.D.Roosevelt teaching Hospital 64

Slovakia Nove Zámky Faculty Hospital Nove Zamky 47

Slovenia Ljubljana Institute of Oncology Ljubljana 64

Slovenia Ljubljana University Medical Centre Ljubljana 108

Spain Barcelona Hospital Sant Pau 73

Spain Barcelona Hospital Universitari Germans Trias I Pujol 103

Spain Pamplona University of Navarra 90

Spain Sabadell Corporacion Sanitaria Parc Tauli 66

Spain Valencia Consorcio Hospital General Universitario de Valencia 61

Spain Valencia Hospital Clinico Valencia 62

Spain Valladolid Hospital Universitario Rio Hortega 82

Sweden Kristianstad Central Hospital in Kristianstad 83

Turkey Ankara Ufuk University Hospital Ankara 30

Turkey Antalya Akdeniz University Hospital 165

Turkey Istanbul Istanbul university, Istanbul medical faculty 81

Turkey Istanbul Acibadem University 25

LAS VEGAS study

Chap

ter

5

123

Country City Institution # Patients

Turkey Istanbul Maltepe University 37

Turkey Izmir Dokuz Eylül Universitesi Tip Fakültesi 161

Turkey Izmir Sifa University Hospital / İzmir 74

Turkey Konya Selcuk University faculty of medicine 138

Turkey Istanbul Fatih Sultan Mehmet Eğitim Ve Araştirma Hastanesi 53

Ukraine Kiev Institute Of Surgery And Transplantology 17

Ukraine Zaporizhzhia Zaporizhzhia State Medical University 18

United Kingdom Barnstaple Northern Devon Healthcare NHS Trust 69

United Kingdom Clydebank, Scotland Golden Jubilee National Hospital 67

United Kingdom Darlington Darlington Memorial Hospital, County Durham and Darlington Foundation NHS Trust 54

United Kingdom Derby Royal Derby Hospital 59

United Kingdom Dorchester Dorset County Hospital 63

United Kingdom Essex The Princess Alexandra NHS Hospital Trust 71

United Kingdom Exeter Royal Devon and Exeter NHS Foundation Trust 123

United Kingdom Great Yarmouth Hospital James Paget University Hospital NHS Foundation Trust 43

United Kingdom Guildford Royal Surrey County Hospital NHS Foundation Trust 70

United Kingdom Kettering Kettering General Hospital NHS Foundation Trust 40

United Kingdom London Barts Health NHS Trust, Royal London Hospital 104

United Kingdom Newcastle upon Tyne

Newcastle Upon Tyne Hospitals NHS Trust The Freeman Hospital High Heaton 125

United Kingdom Plymouth Derriford Hospital Plymouth Hospitals NHS Trust 179

United Kingdom Sheffield Royal Hallamshire Hospital 103

United Kingdom Stafford Mid Staffordshire NHS 28

United Kingdom Taunton Musgrove Park Hospital 50

United Kingdom Torquay, Torbay South Devon Healthcare NHS Foundation Trust /Torbay Hospital, 83

United Kingdom Truro Royal Cornwall Hospital 81

United Kingdom Wakefield Mid Yorkshire Hospitals NHS Trust; Pinderfields Hospital 65

United Kingdom West Bromich Sandwell and West Birmingham NHS Trust 127

124

Country City Institution # Patients

United Kingdom York York Teaching Hospitals NHS Foundation Trust 81

United States Aurora, Colorado University of Colorado School of Medicine/University of Colorado Hospital 56

United States Boston Massachusetts General Hospital, Boston 120

United States Rochester Mayo Clinic 200

eTable 5. Hospital characteristics of participating centres

Centres responded (N = 145)

Community hospital 33

Teaching hospital 108

Number of hospital beds 600 [400 – 960]

Number of ICU beds 20 [11 – 35]

Number of operating theatres 15 [9 -22]

Staff (anaesthesiologists) 25 [12 – 40]

Invasive mechanical ventilation for surgery per week 103 [60 – 200]

Surgical procedures

Abdominal surgery 135 (93%)

Bariatric surgery 66 (46%)

Cardiothoracic surgery 71 (49%)

Endocrine surgery 88 (61%)

Eye surgery 97 (67%)

General surgery 132 (91%)

Gynaecological surgery 126 (87%)

Interventional neuroradiology 56 (39%)

Neurosurgery 82 (57%)

Oral and maxillofacial surgery 96 (62%)

Orthopaedic surgery 121 (83%)

Plastic surgery 91 (63%)

Surgical oncology 107 (74%)

Trauma surgery 111 (77%)

Urology

Vascular surgery

Data are presented as median [QR] or N (%). One participating centre did not respond; ICU: intensive care unit

LAS VEGAS study

Chap

ter

5

125

eTab

le 6

. Dem

ogra

phic

s an

d su

rgic

al c

hara

cter

istic

s

Varia

ble

All p

atien

tsBM

I <35

BMI ≥

35

Non

lapa

rosc

opic

Lapa

rosc

opic

ARIS

CAT

< 26

ARIS

CAT

≥ 26

Ethn

icity

Ca

ucas

ian

91.2

(749

3/82

15)

91.1

(681

9/74

86)

92.3

(656

/711

)91

.7 (6

065/

6614

)89

.2 (1

428/

1601

)90

.7 (5

511/

6079

)92

.8 (1

982/

2136

)

Bl

ack

ethn

icity

1.1

(87/

8215

)1.

1 (8

0/74

86)

1.0

(7/7

11)

1.1

(71/

6614

)1.

0 (1

6/16

01)

1.2

(73/

6079

)0.

7 (1

4/21

36)

As

ian

2.5

(206

/821

5)2.

5 (1

90/7

486)

2.3

(16/

711)

2.5

(163

/661

4)2.

7 (4

3/16

01)

2.5

(152

/607

9)2.

5 (5

4/21

36)

O

ther

5.3

(429

/821

5)5.

3 (3

97/7

486)

4.5

(32/

711)

4.8

(315

/661

4)7.

1 (1

14/1

601)

5.6

(343

/607

9)4.

0 (8

6/21

36)

Smok

ing

(cur

rent

) 23

.6 (1

943/

8241

)24

.2 (1

815/

7507

)17

.6 (1

26/7

16)

24.2

(160

9/66

36)

20.8

(334

/160

5)24

.8 (1

515/

6097

)20

.0 (4

28/2

144)

Resp

irato

ry

infe

ction

(< 3

0 da

ys

preo

pera

tivel

y)3.

8 (3

10/8

241)

3.7

(281

/750

7)4.

1 (2

9/71

6)3.

8 (2

53/6

636)

3.6

(57/

1605

)1.

8 (1

10/6

097)

9.3

(200

/214

4)

Whi

te b

lood

cel

ls (x

109 /L

)*7.

0 (5

.9 –

8.7

)7.

0 (5

.7 –

8.5

)7.

8 (6

.0 –

9.0

)7.

0 (5

.8 –

8.6

)7.

0 (5

.9 –

8.9

)7.

0 (5

.8 –

8.3

)7.

0 (5

.9 –

9.0

)

< 4

2.6

(169

/654

0)2.

7 (1

64/5

978)

0.9

(5/5

47)

2.6

(137

/523

7)2.

5 (3

2/13

03)

2.4

(110

/465

4)3.

1 (5

9/18

86)

4 –

11

89.9

(588

1/65

40)

90.0

(537

9/59

78)

89.8

(491

/547

)90

.1 (4

720/

5237

)89

.1 (1

161/

1303

)90

.8 (4

227/

4654

)87

.7 (1

654/

1886

)

> 1

17.

5 (4

90/6

540)

7.3

(435

/597

8)9.

3 (5

1/54

7)7.

3 (3

80/5

237)

8.4

(110

/130

3)6.

8 (3

17/4

654)

9.2

(173

/188

6)

Crea

tinin

e –

mg/

dL*

0.80

(0.7

0 –

0.99

)0.

81 (0

.70

– 0.

99)

0.80

(0.7

0 –

0.97

)0.

81 (0

.70

– 1.

0)0.

80 (0

.70

– 0.

95)

0.80

(0.7

0 –

0.96

)0.

84 (0

.70

– 1.

05)

< 1

.595

.3 (5

747/

6032

)95

.2 (5

238/

5500

)95

.9 (4

95/5

16)

94.7

(456

4/48

19)

97.5

(118

3/12

13)

96.7

(405

4/41

92)

92.0

(169

3/18

40)

1.5

– 3

.03.

2 (1

95/6

032)

3.2

(174

/550

0)3.

7 (1

9/51

6)3.

7 (1

77/4

819)

1.5

(18/

1213

)2.

3 (9

8/41

92)

5.3

(97/

1840

)

> 3

.01.

5 (9

0/60

32)

1.6

(88/

5500

)0.

4 (2

/516

)1.

6 (7

8/48

19)

1.0

(12/

1213

)1.

0 (4

0/41

92)

2.7

(50/

1840

)

Antib

iotic

pro

phyl

axis

68.8

(566

7/82

41)

68.3

(512

7/75

07)

73.5

(526

/716

)68

.2 (4

524/

6636

)71

.2 (1

143/

1605

)61

.9 (3

777/

6097

)88

.2 (1

890/

2144

)

Data

is p

rese

nted

as:

med

ian

(QR)

or p

ropo

rtion

(n/N

); *L

abor

ator

y va

lues

: whi

te b

lood

cel

l cou

nt a

nd c

reati

nine

wer

e co

llect

ed w

hen

avai

labl

e fr

om c

olle

ction

with

in ro

utine

car

e

126

eTable 7. Univariate and Multivariate Logistic Regression in all patients (n = 8241)

Variables Univariate ModelOR (95% CI), p value

Multivariate ModelOR (95% CI), p value

Ventilation parameters

Tidal volume, ml/kg PBW 1.00 (0.96 – 1.05), 0.737 ---

PEEP, cmH2O 1.14 (1.11 – 1.18), < 0.0001 1.06 (1.01 – 1.12), 0.020

Peak pressure, cmH2O 1.06 (1.04 – 1.07), < 0.0001 1.03 (1.00 – 1.06), 0.040

FiO2 0.99 (0.99 – 0.99), < 0.0001 1.00 (0.99 – 1.01), 0.885

Characteristics of patients

Male sex 1.11 (0.96 – 1.27), 0.163 0.99 (0.79 – 1.25), 0.966

BMI, kg/m2 1.02 (1.01 – 1.03), < 0.0001 1.01 (0.99 – 1.03), 0.081

COPD 1.89 (1.48 – 2.40), < 0.0001 1.34 (0.97 – 1.86), 0.088

Age, years ≤ 50 51 – 80 > 80

1 (Reference)2.21 (1.89 – 2.60), < 0.00014.32 (3.19 – 5.84), < 0.0001

1 (Reference)1.96 (1.50 – 2.59), < 0.00013.12 (1.79 – 5.42), < 0.0001

Functional Status Independent Partially Dependent Totally Dependent

1 (Reference)2.65 (2.10 – 3.32), < 0.00012.13 (1.25 – 3.64), 0.005

1 (Reference)1.54 (1.00 – 2.35), 0.0471.93 (0.95 – 3.92), 0.069

Current Smoker 0.73 (0.61 – 0.87), 0.001 0.83 (0.65 – 1.05), 0.120

Respiratory Infection < 30 days 1.77 (1.31 – 2.41), < 0.0001 1.68 (1.09 – 2.58), 0.019

Preoperative SpO2, % ≥ 96 91 – 95 ≤ 90

1 (Reference)2.00 (1.67 – 2.40), < 0.00016.00 (3.83 – 9.38), < 0.0001

1 (Reference)1.39 (1.10 – 1.76), 0.0052.55 (1.26 – 5.15), 0.009

Perioperative anaemia (Hb < 10 g/dl) 2.14 (1.69 – 2.72), < 0.0001 1.92 (1.18 – 3.14), 0.009

Characteristics of surgery

Non-Laparoscopic surgery 1.10 (0.92 – 1.32), 0.288 ---

Moment of Surgery Elective Urgency Emergency

1 (Reference)2.10 (1.68 – 2.62), < 0.00012.87 (1.97 – 4.18), < 0.0001

1 (Reference)1.55 (1.09 – 2.21), 0.0142.63 (1.44 – 4.82), 0.002

Planned Duration of Surgery, hours ≤ 2 2 – 3 > 3

1 (Reference)2.21 (1.86 – 2.62), < 0.00013.68 (3.06 – 4.42), < 0.0001

1 (Reference)1.44 (1.13 – 1.83), 0.0032.16 (1.60 – 2.91), < 0.0001

Type of Incision Peripheral Abdominal

1 (Reference)1.87 (1.62 – 2.15), < 0.0001

1 (Reference)1.30 (0.96 – 1.75), 0.091

LAS VEGAS study

Chap

ter

5

127

Intraoperative characteristics

Fluid intake, millilitres < 999 1000 – 1999 2000 – 2999 ≥ 3000

1 (Reference)1.28 (1.04 – 1.56), 0.0172.67 (2.13 – 3.36), < 0.00014.25 (3.34 – 5.41), < 0.0001

1 (Reference)1.58 (1.13 – 2.23), 0.0092.19 (1.41 – 3.42), 0.0012.93 (1.78 – 4.82), < 0.0001

Epidural anaesthesia 3.14 (2.49 – 3.96), < 0.0001 1.22 (0.87 – 1.71), 0.253

No use of neuromuscular blocking agents 1.87 (1.45 – 2.36), < 0.0001 0.85 (0.58 – 1.23), 0.387

Desaturation 3.75 (2.92 – 4.82), < 0.0001 2.23 (1.41 – 3.54), < 0.0001

Arrhythmia 3.57 (1.91 – 6.68), < 0.0001 2.01 (0.78 – 5.22), 0.149

Hypotension 2.13 (1.84 – 2.46), < 0.0001 1.04 (0.73 – 1.47), 0.833

Need of vasoactive drugs 2.66 (2.29 – 3.10), < 0.0001 1.08 (0.72 – 1.62), 0.691

Postoperative characteristics

No reversal of neuromuscular blockade (0%) 1.22 (1.05 – 1.42), 0.008 1.32 (0.99 – 1.75), 0.054

Postoperative residual curarisation 6.32 (3.75 – 10.65), < 0.0001 3.39 (1.57 – 7.29), 0.002

Odds ratio according to one unit increase of variable; PBW, predicted body weight; PEEP, positive end-expiratory pressure; Peak pressure: peak inspiratory pressure; FiO2, inspired fraction of oxygen; BMI: body mass index; COPD: chronic obstructive pulmonary disease; SpO2: peripheral oxygen saturation; Hb: haemoglobin

eTable 8. Univariate and multivariate analyses of overall cohort using categories of PEEP and tidal volume (adjusted by the same co-variates describing for the overall cohort in table 5)

Variable Univariate Analysis OR (95% CI), p value

Multivariable Analysis OR (95% CI), p value

PEEP, cmH2O 0 – 2 3 – 6 ≥ 7

1 (Reference)1.53 (1.31 – 1.79), < 0.0013.21 (2.46 – 4.19), < 0.001

1 (Reference)1.30 (0.99 – 1.71), 0.0611.56 (1.02 – 2.52), 0.033

Tidal volume, ml/kg PBW < 7 7 – 9 ≥ 10

1 (Reference)0.89 (0.74 – 1.07), 0.2061.05 (0.82 – 1.35), 0.704

1 (Reference)0.87 (0.66 – 1.14), 0.3070.86 (0.59 – 1.26), 0.449

PEEP: positive end-expiratory pressure; PBW: predicted body weight; OR: odds ratio; CI: confidence interval

128

eTab

le 9

. Pati

ents

cha

ract

eris

tics

and

surg

ery

char

acte

ristic

s aft

er m

atch

ing

on p

reop

erati

ve ri

sk o

f PPC

sa

Varia

bles

Unm

atch

ed C

ohor

t (n

= 82

41)

Stan

dard

ized

D

iffer

ence

, %b

(p v

alue

c )

Mat

ched

Coh

ort (

n =

2481

)St

anda

rdiz

ed

Diff

eren

ce, %

b (p

val

uec )

PPC

(n =

861

)N

o PP

C(n

= 7

380)

PPC

(n =

546

)N

o PP

C(n

= 1

405)

Base

line

char

acte

ristic

s of

pati

ents

Age,

yea

rs62

.0 (4

9.0

– 72

.0)

53.0

(39.

0 –

65.0

)49

.1 (<

0.0

001)

63.0

(51.

0 –

73.0

)64

.0 (5

2.0

– 72

.0)

- 3.8

(0.4

52)

Mal

e se

x40

7 (4

7.3)

3304

(44.

8) 5

.0 (0

.162

)25

7 (4

7.1)

660

(47.

0)0.

2 (0

.970

)

BMI,

kg/m

226

.8 (2

3.8

– 30

.5)

26.2

(23.

3 –

29.9

)13

.0 (<

0.0

001)

27.0

(24.

0 –

31.2

) 27

.3 (2

4.1

– 31

.1)

- 1.8

(0.5

15)

ARIS

CAT

scor

e26

.0 (1

4.5

– 40

.5)

15.0

(3.0

– 2

3.0)

69

.3 (<

0.0

001)

26.0

(15.

0 –

40.0

) 26

.0 (1

6.0

– 38

.0)

0.3

(0.9

88)

Preo

pera

tive

SpO

2, %

97.0

(95.

0 –

98.0

)98

.0 (9

6.0

– 99

.0)

- 39.

2 (<

0.0

001)

97.0

(95.

0 –

98.0

) 97

.0 (9

5.0

– 98

.0)

- 0.4

(0.4

26)

Func

tiona

l sta

tus

In

depe

nden

t

Parti

ally

dep

ende

nt

Tota

lly d

epen

dent

736

(85.

7)10

6 (1

2.3)

17 (2

.0)

6926

(93.

9)37

7 (5

.1)

75 (1

.0)

- 45

.6 (<

0.0

001)

473

(86.

6)66

(12.

1)7

(1.3

)

1231

(87.

6)15

6 (1

1.1)

18 (1

.3)

- 4.4

(0.8

27)

Smok

er, c

urre

nt16

2 (1

8.8)

1781

(24.

1)- 1

5.8

(0.0

01)

95 (1

7.4)

234

(16.

7)2.

5 (0

.0.6

93)

COPD

87 (1

0.1)

415

(5.6

)31

.3 (<

0.0

001)

54 (9

.9)

150

(10.

7)- 4

.5 (0

.610

)

Chro

nic

co-m

orbi

dity

313

(36.

4)14

47 (1

9.6)

43.2

(< 0

.000

1)19

6 (3

5.9)

515

(36.

7)- 1

.7 (0

.755

)

Resp

irato

ry in

fecti

on<3

0 da

ys52

(6.0

)25

8 (3

.5)

27.7

(< 0

.000

1)38

(7.0

)93

(6.6

)3.

1 (0

.787

)

Perio

pera

tive

anae

mia

56 (6

.5)

182

(2.5

)47

.0 (<

0.0

001)

63 (1

1.5)

148

(10.

5)5.

1 (0

.521

)

Char

acte

ristic

s of s

urge

ry

Lapa

rosc

opic

surg

ery

156

(18.

1)14

49 (1

9.6)

- 4.9

(0.2

87)

107

(19.

6)28

2 (2

0.1)

- 1.6

(0.8

13)

Plan

ned

dura

tion

of

surg

ery

2 ho

urs

2

– 3

hour

s

> 3

hour

s

418

(48.

5)23

5 (2

7.3)

208

(24.

2)

5306

(72.

0)13

49 (1

8.3)

717

(9.7

)

- 52.

1 (<

0.0

001)

267

(48.

9)15

5 (2

8.4)

124

(22.

7)

669

(47.

6)41

6 (2

9.6)

320

(22.

8)

2.6

(0.8

45)

Cond

ition

of s

urge

ry

Elec

tive

U

rgen

cy

Emer

genc

y

716

(83.

2)10

8 (1

2.5)

37 (4

.3)

6770

(91.

7)48

7 (6

.6)

122

(1.7

)

- 39.

4 (<

0.0

001)

461

(84.

4)68

(12.

5)17

(3.1

)

1211

(86.

2)15

7 (1

1.2)

37 (2

.6)

- 7.2

(0.5

98)

Type

of i

ncisi

on

Perip

hera

l

Abdo

min

al42

9 (4

9.8)

432

(50.

2)47

94 (6

5.0)

2586

(35.

0)- 3

1.8

(< 0

.000

1)25

8 (4

7.3)

288

(52.

7)66

1 (4

7.0)

744

(53.

0)0.

6 (0

.934

)

Flui

d in

take

, mill

ilitr

es15

00 (1

000

– 25

00)

1000

(750

– 1

500)

44.6

(< 0

.000

1)15

00 (1

000

– 23

00)

1500

(100

0 –

2200

)5.

7 (0

.074

)

Epid

ural

ana

esth

esia

106

(12.

3)31

6 (4

.3)

53.7

(< 0

.000

1)67

(12.

3)17

6 (1

2.5)

- 0.9

(0.8

77)

Use

of N

MBA

775

(90.

0)61

15 (8

3.0)

30.3

(< 0

.000

1)50

2 (9

1.9)

1266

(90.

1)11

.0 (0

.212

)

Reve

rsal

of N

MBA

288

(33.

5)28

09 (3

8.2)

- 10.

2 (0

.008

)18

4 (3

3.7)

430

(30.

6)7.

1 (0

.186

)

Resid

ual c

urar

isatio

n24

(3.2

)36

(0.5

)75

.1 (<

0.0

001)

11 (2

.0)

27 (1

.9)

2.6

(0.8

93)

Intr

aope

rativ

e co

mpl

icati

ons

Desa

tura

tion

95 (1

1.0)

236

(3.2

)60

.5 (<

0.0

001)

53 (9

.7)

55 (3

.9)

46.4

(< 0

.000

1)

Hypo

tens

ion

364

(42.

3)18

87 (2

5.6)

38.4

(< 0

.000

1)22

1 (4

0.5)

475

(33.

8)14

.4 (0

.005

)

Arrh

ythm

ias

14 (1

.6)

34 (0

.5)

53.1

(< 0

.000

1)9

(1.6

)10

(0.7

)39

.6 (0

.005

)

Nee

d of

vas

oacti

ve d

rugs

361

(42.

0)15

77 (2

1.4)

50.0

(< 0

.000

1)21

0 (3

8.5)

448

(31.

9)14

.5 (0

.058

)

Venti

lato

ry p

aram

eter

s

PEEP

, cm

H2O

5.0

(2.0

– 5

.0)

4.0

(0.0

– 5

.0)

31.3

(< 0

.001

)5.

0 (2

.0 –

5.0

)4.

0 (1

.0 –

5.0

)19

.4 (<

0.0

01)

Tida

l vol

ume,

ml/k

g PB

W8.

0 (7

.2 –

9.1

)8.

1 (7

.2 –

9.1

)- 0

.6 (0

.417

)8.

1 (7

.2 –

9.2

)8.

3 (7

.4 –

9.3

)- 7

.2 (0

.030

)

Peak

pre

ssur

e, c

mH 2

O18

.5 (1

6.0

– 22

.0)

17.0

(15.

0 –

20.5

)28

.0 (<

0.0

01)

19.0

(16.

0 –

23.0

)18

.0 (1

6.0

– 21

.0)

10.3

(0.1

17)

FiO

20.

50 (0

.45

– 0.

64)

0.51

(0.4

5 –

0.70

)- 1

3.3

(0.0

04)

0.50

(0.4

6 –

0.62

)0.

50 (0

.45

– 0.

65)

- 2.6

(0.9

11)

PPC,

pos

tope

rativ

e pu

lmon

ary

com

plic

ation

s; B

MI,

body

mas

s ind

ex; A

RISC

AT, T

he A

sses

s Res

pira

tory

Risk

in S

urgi

cal P

atien

ts in

Cat

alon

ia; S

pO2:

per

iphe

ral o

xyge

n sa

tura

tion;

CO

PD,

chro

nic

obst

ructi

ve p

ulm

onar

y di

seas

e; M

IN, m

inut

es; P

RBC,

pac

ked

red

bloo

d ce

lls; N

MBA

, neu

rom

uscu

lar b

lock

ing

agen

ta)

Val

ues a

re e

xpre

ssed

as n

umbe

r (pe

rcen

tage

) or m

edia

n (Q

R)b)

Indi

cate

s the

diff

eren

ce b

etw

een

the

mea

ns fo

r the

two

grou

ps d

ivid

ed b

y th

e m

utua

l sta

ndar

d de

viati

onc)

Com

paris

on o

f diff

eren

ces b

etw

een

the

two

grou

ps u

sing

the

t tes

t for

con

tinuo

us v

aria

bles

and

the

χ2 te

st fo

r cat

egor

ical

var

iabl

es

LAS VEGAS study

Chap

ter

5

129

eTab

le 9

. Pati

ents

cha

ract

eris

tics

and

surg

ery

char

acte

ristic

s aft

er m

atch

ing

on p

reop

erati

ve ri

sk o

f PPC

sa

Varia

bles

Unm

atch

ed C

ohor

t (n

= 82

41)

Stan

dard

ized

D

iffer

ence

, %b

(p v

alue

c )

Mat

ched

Coh

ort (

n =

2481

)St

anda

rdiz

ed

Diff

eren

ce, %

b (p

val

uec )

PPC

(n =

861

)N

o PP

C(n

= 7

380)

PPC

(n =

546

)N

o PP

C(n

= 1

405)

Base

line

char

acte

ristic

s of

pati

ents

Age,

yea

rs62

.0 (4

9.0

– 72

.0)

53.0

(39.

0 –

65.0

)49

.1 (<

0.0

001)

63.0

(51.

0 –

73.0

)64

.0 (5

2.0

– 72

.0)

- 3.8

(0.4

52)

Mal

e se

x40

7 (4

7.3)

3304

(44.

8) 5

.0 (0

.162

)25

7 (4

7.1)

660

(47.

0)0.

2 (0

.970

)

BMI,

kg/m

226

.8 (2

3.8

– 30

.5)

26.2

(23.

3 –

29.9

)13

.0 (<

0.0

001)

27.0

(24.

0 –

31.2

) 27

.3 (2

4.1

– 31

.1)

- 1.8

(0.5

15)

ARIS

CAT

scor

e26

.0 (1

4.5

– 40

.5)

15.0

(3.0

– 2

3.0)

69

.3 (<

0.0

001)

26.0

(15.

0 –

40.0

) 26

.0 (1

6.0

– 38

.0)

0.3

(0.9

88)

Preo

pera

tive

SpO

2, %

97.0

(95.

0 –

98.0

)98

.0 (9

6.0

– 99

.0)

- 39.

2 (<

0.0

001)

97.0

(95.

0 –

98.0

) 97

.0 (9

5.0

– 98

.0)

- 0.4

(0.4

26)

Func

tiona

l sta

tus

In

depe

nden

t

Parti

ally

dep

ende

nt

Tota

lly d

epen

dent

736

(85.

7)10

6 (1

2.3)

17 (2

.0)

6926

(93.

9)37

7 (5

.1)

75 (1

.0)

- 45

.6 (<

0.0

001)

473

(86.

6)66

(12.

1)7

(1.3

)

1231

(87.

6)15

6 (1

1.1)

18 (1

.3)

- 4.4

(0.8

27)

Smok

er, c

urre

nt16

2 (1

8.8)

1781

(24.

1)- 1

5.8

(0.0

01)

95 (1

7.4)

234

(16.

7)2.

5 (0

.0.6

93)

COPD

87 (1

0.1)

415

(5.6

)31

.3 (<

0.0

001)

54 (9

.9)

150

(10.

7)- 4

.5 (0

.610

)

Chro

nic

co-m

orbi

dity

313

(36.

4)14

47 (1

9.6)

43.2

(< 0

.000

1)19

6 (3

5.9)

515

(36.

7)- 1

.7 (0

.755

)

Resp

irato

ry in

fecti

on<3

0 da

ys52

(6.0

)25

8 (3

.5)

27.7

(< 0

.000

1)38

(7.0

)93

(6.6

)3.

1 (0

.787

)

Perio

pera

tive

anae

mia

56 (6

.5)

182

(2.5

)47

.0 (<

0.0

001)

63 (1

1.5)

148

(10.

5)5.

1 (0

.521

)

Char

acte

ristic

s of s

urge

ry

Lapa

rosc

opic

surg

ery

156

(18.

1)14

49 (1

9.6)

- 4.9

(0.2

87)

107

(19.

6)28

2 (2

0.1)

- 1.6

(0.8

13)

Plan

ned

dura

tion

of

surg

ery

2 ho

urs

2

– 3

hour

s

> 3

hour

s

418

(48.

5)23

5 (2

7.3)

208

(24.

2)

5306

(72.

0)13

49 (1

8.3)

717

(9.7

)

- 52.

1 (<

0.0

001)

267

(48.

9)15

5 (2

8.4)

124

(22.

7)

669

(47.

6)41

6 (2

9.6)

320

(22.

8)

2.6

(0.8

45)

Cond

ition

of s

urge

ry

Elec

tive

U

rgen

cy

Emer

genc

y

716

(83.

2)10

8 (1

2.5)

37 (4

.3)

6770

(91.

7)48

7 (6

.6)

122

(1.7

)

- 39.

4 (<

0.0

001)

461

(84.

4)68

(12.

5)17

(3.1

)

1211

(86.

2)15

7 (1

1.2)

37 (2

.6)

- 7.2

(0.5

98)

Type

of i

ncisi

on

Perip

hera

l

Abdo

min

al42

9 (4

9.8)

432

(50.

2)47

94 (6

5.0)

2586

(35.

0)- 3

1.8

(< 0

.000

1)25

8 (4

7.3)

288

(52.

7)66

1 (4

7.0)

744

(53.

0)0.

6 (0

.934

)

Flui

d in

take

, mill

ilitr

es15

00 (1

000

– 25

00)

1000

(750

– 1

500)

44.6

(< 0

.000

1)15

00 (1

000

– 23

00)

1500

(100

0 –

2200

)5.

7 (0

.074

)

Epid

ural

ana

esth

esia

106

(12.

3)31

6 (4

.3)

53.7

(< 0

.000

1)67

(12.

3)17

6 (1

2.5)

- 0.9

(0.8

77)

Use

of N

MBA

775

(90.

0)61

15 (8

3.0)

30.3

(< 0

.000

1)50

2 (9

1.9)

1266

(90.

1)11

.0 (0

.212

)

Reve

rsal

of N

MBA

288

(33.

5)28

09 (3

8.2)

- 10.

2 (0

.008

)18

4 (3

3.7)

430

(30.

6)7.

1 (0

.186

)

Resid

ual c

urar

isatio

n24

(3.2

)36

(0.5

)75

.1 (<

0.0

001)

11 (2

.0)

27 (1

.9)

2.6

(0.8

93)

Intr

aope

rativ

e co

mpl

icati

ons

Desa

tura

tion

95 (1

1.0)

236

(3.2

)60

.5 (<

0.0

001)

53 (9

.7)

55 (3

.9)

46.4

(< 0

.000

1)

Hypo

tens

ion

364

(42.

3)18

87 (2

5.6)

38.4

(< 0

.000

1)22

1 (4

0.5)

475

(33.

8)14

.4 (0

.005

)

Arrh

ythm

ias

14 (1

.6)

34 (0

.5)

53.1

(< 0

.000

1)9

(1.6

)10

(0.7

)39

.6 (0

.005

)

Nee

d of

vas

oacti

ve d

rugs

361

(42.

0)15

77 (2

1.4)

50.0

(< 0

.000

1)21

0 (3

8.5)

448

(31.

9)14

.5 (0

.058

)

Venti

lato

ry p

aram

eter

s

PEEP

, cm

H2O

5.0

(2.0

– 5

.0)

4.0

(0.0

– 5

.0)

31.3

(< 0

.001

)5.

0 (2

.0 –

5.0

)4.

0 (1

.0 –

5.0

)19

.4 (<

0.0

01)

Tida

l vol

ume,

ml/k

g PB

W8.

0 (7

.2 –

9.1

)8.

1 (7

.2 –

9.1

)- 0

.6 (0

.417

)8.

1 (7

.2 –

9.2

)8.

3 (7

.4 –

9.3

)- 7

.2 (0

.030

)

Peak

pre

ssur

e, c

mH 2

O18

.5 (1

6.0

– 22

.0)

17.0

(15.

0 –

20.5

)28

.0 (<

0.0

01)

19.0

(16.

0 –

23.0

)18

.0 (1

6.0

– 21

.0)

10.3

(0.1

17)

FiO

20.

50 (0

.45

– 0.

64)

0.51

(0.4

5 –

0.70

)- 1

3.3

(0.0

04)

0.50

(0.4

6 –

0.62

)0.

50 (0

.45

– 0.

65)

- 2.6

(0.9

11)

PPC,

pos

tope

rativ

e pu

lmon

ary

com

plic

ation

s; B

MI,

body

mas

s ind

ex; A

RISC

AT, T

he A

sses

s Res

pira

tory

Risk

in S

urgi

cal P

atien

ts in

Cat

alon

ia; S

pO2:

per

iphe

ral o

xyge

n sa

tura

tion;

CO

PD,

chro

nic

obst

ructi

ve p

ulm

onar

y di

seas

e; M

IN, m

inut

es; P

RBC,

pac

ked

red

bloo

d ce

lls; N

MBA

, neu

rom

uscu

lar b

lock

ing

agen

ta)

Val

ues a

re e

xpre

ssed

as n

umbe

r (pe

rcen

tage

) or m

edia

n (Q

R)b)

Indi

cate

s the

diff

eren

ce b

etw

een

the

mea

ns fo

r the

two

grou

ps d

ivid

ed b

y th

e m

utua

l sta

ndar

d de

viati

onc)

Com

paris

on o

f diff

eren

ces b

etw

een

the

two

grou

ps u

sing

the

t tes

t for

con

tinuo

us v

aria

bles

and

the

χ2 te

st fo

r cat

egor

ical

var

iabl

es

130

eTable 10. Univariate and Multivariate Logistic Regression in Patients with BMI ≥ 35 kg/m2 (n = 716)

Variables Univariate ModelOR (95% CI), p value

Multivariate ModelOR (95% CI), p value

Ventilation parameters

Tidal volume, ml/kg PBW 1.10 (0.99 – 1.21), 0.067 0.94 (0.81 – 1.08), 0.387

PEEP, cmH2O 1.18 (1.09 – 1.28), < 0.0001 1.13 (1.01 – 1.26), 0.032

Peak pressure, cmH2O 1.10 (1.05 – 1.15), < 0.0001 1.09 (1.03 – 1.15), 0.003

FiO2 0.99 (0.98 – 1.01), 0.380 ---

Characteristics of patients

Male sex 1.13 (0.72 – 1.77), 0.600 ---

COPD 0.94 (0.41 – 2.15), 0.890 ---

Age, years ≤ 50 51 – 80 > 80

1 (Reference)1.74 (1.11 – 2.73), 0.0162.35 (0.62 – 8.88), 0.207

1 (Reference)1.89 (1.03 – 3.47), 0.0392.26 (0.33 – 15.59), 0.406

Functional Status Independent Partially Dependent Totally Dependent

1 (Reference)1.11 (0.51 – 2.43), 0.7930.68 (0.08 – 5.42), 0.715

---

Current Smoker 1.02 (0.59 – 1.76), 0.949 ---

Respiratory Infection < 30 days 2.91 (1.29 – 6.59), 0.010 1.84 (0.77 – 4.42), 0.170

Preoperative SpO2, % ≥ 96 91 – 95 ≤ 90

1 (Reference)1.64 (1.00 – 2.69), 0.0496.18 (2.00 – 19.02), 0.002

1 (Reference)1.01 (0.54 – 1.89), 0.9693.09 (0.86 – 11.01), 0.082

Perioperative anaemia (Hb < 10 g/dl) 1.45 (0.57 – 3.67), 0.432 ---

Characteristics of surgery

Non-Laparoscopic surgery 0.76 (0.48 – 1.18), 0.219 ---

Moment of Surgery Elective Urgency Emergency

1 (Reference)2.03 (0.96 – 4.27), 0.0622.60 (0.50 – 13.59), 0.258

1 (Reference)1.26 (0.48 – 3.33), 0.6372.21 (0.37 – 13.03), 0.379

Planned Duration of Surgery, hours ≤ 2 2 – 3 > 3

1 (Reference)2.08 (1.29 – 3.37), 0.0032.81 (1.55 – 5.11), 0.001

1 (Reference)1.14 (0.67 – 1.93), 0.6301.31 (0.55 – 3.12), 0.535

Type of Incision Peripheral Abdominal

1 (Reference)2.04 (1.33 – 3.15), 0.001

1 (Reference)1.38 (0.79 – 2.41), 0.262

LAS VEGAS study

Chap

ter

5

131

Intraoperative characteristics

Fluid intake, millilitres < 999 1000 – 1999 2000 – 2999 ≥ 3000

1 (Reference)1.28 (0.68 – 2.41), 0.4502.68 (1.29 – 5.56), 0.0084.29 (2.00 – 9.15), < 0.0001

1 (Reference)0.99 (0.47 – 2.09), 0.9921.79 (0.68 – 4.74), 0.2393.19 (1.10 – 9.29), 0.033

Epidural anaesthesia 3.81 (1.81 – 8.02), < 0.0001 1.84 (0.57 – 6.01), 0.309

No use of neuromuscular blocking agents 2.27 (0.96 – 5.37), 0.061 0.74 (0.22 – 2.54), 0.632

Desaturation 4.11 (2.42 – 6.96), < 0.0001 3.34 (1.59 – 7.02), 0.001

Arrhythmia 3.05 (0.27 – 34.00), 0.364 ---

Hypotension 2.17 (1.41 – 3.35), < 0.0001 1.54 (0.74 – 3.24), 0.250

Need of vasoactive drugs 2.04 (1.31 – 3.18), 0.002 0.62 (0.26 – 1.51), 0.291

Postoperative characteristics

No reversal of neuromuscular blockade 1.03 (0.67 – 1.57), 0.905 ---

Postoperative residual curarisation 6.36 (1.26 – 32.00), 0.025 4.98 (0.94 – 26.46), 0.059

Odds ratio according to one unit increase of variable; PBW, predicted body weight; PEEP, positive end-expiratory pressure; Peak pressure: peak inspiratory pressure; FiO2, inspired fraction of oxygen; BMI: body mass index; COPD: chronic obstructive pulmonary disease; SpO2: peripheral oxygen saturation; Hb: haemoglobin

132

eTable 11. Univariate and Multivariate Logistic Regression in Patients Under Laparoscopic Surgery (n = 1605)

Variables Univariate ModelOR (95% CI), p value

Multivariate ModelOR (95% CI), p value

Ventilation parameters

Tidal volume, ml/kg PBW 1.11 (1.00 – 1.22), 0.051 1.05 (0.91 – 1.22), 0.476

PEEP, cmH2O 1.12 (1.04 – 1.20), 0.002 1.10 (0.99 – 1.22), 0.058

Peak pressure, cmH2O 1.08 (1.05 – 1.12), < 0.0001 1.01 (0.96 – 1.07), 0.621

FiO2 0.99 (0.97 – 0.99), 0.012 1.00 (0.99 – 1.01), 0.996

Characteristics of patients

Male sex 1.21 (0.86 – 1.70), 0.276 ---

BMI, kg/m2 1.04 (1.02 – 1.07), < 0.0001 1.03 (0.99 – 1.06), 0.074

COPD 1.64 (0.89 – 3.03), 0.113 0.90 (0.34 – 2.38), 0.833

Age, years ≤ 50 51 – 80 > 80

1 (Reference)2.40 (1.68 – 3.43), < 0.00013.56 (1.57 – 8.08), 0.002

1 (Reference)2.00 (1.24 – 3.23), 0.0041.37 (0.49 – 3.82), 0.544

Functional Status Independent Partially Dependent Totally Dependent

1 (Reference)2.79 (1.44 – 5.42), 0.0021.08 (0.14 – 8.63), 0.938

1 (Reference)1.65 (0.58 – 4.70), 0.3524.51 (0.32 – 63.48), 0.264

Current Smoker 0.82 (0.53 – 1.25), 0.355 ---

Respiratory Infection < 30 days 2.04 (1.01 – 4.13), 0.047 2.35 (0.98 – 5.62), 0.055

Preoperative SpO2, % ≥ 96 91 – 95 ≤ 90

1 (Reference)2.08 (1.34 – 3.23), 0.0014.40 (1.12 – 17.28), 0.033

1 (Reference)1.57 (0.91 – 2.69), 0.1013.81 (0.47 – 30.91), 0.210

Perioperative anaemia (Hb < 10 g/dl) 2.24 (1.21 – 4.12), 0.010 2.58 (0.76 – 8.74), 0.127

Characteristics of surgery

Moment of Surgery Elective Urgency Emergency

1 (Reference)1.25 (0.71 – 2.20), 0.4391.15 (0.40 – 3.30), 0.792

---

Planned Duration of Surgery, hours ≤ 2 2 – 3 > 3

1 (Reference)2.43 (1.61 – 3.65), < 0.00014.22 (2.72 – 6.55), < 0.0001

1 (Reference)1.89 (0.99 – 3.62), 0.0541.98 (0.91 – 4.32), 0.086

LAS VEGAS study

Chap

ter

5

133

Intraoperative characteristics

Fluid intake, millilitres < 999 1000 – 1999 2000 – 2999 ≥ 3000

1 (Reference)1.04 (0.65 – 1.65), 0.8751.66 (0.96 – 2.88), 0.0693.59 (1.96 – 66.56), < 0.0001

1 (Reference)1.28 (0.63 – 2.62), 0.4900.93 (0.28 – 3.04), 0.9072.46 (0.80 – 7.57), 0.118

Epidural anaesthesia 3.38 (1.76 – 6.48), < 0.0001 2.56 (1.33 – 4.94), 0.005

No use of neuromuscular blocking agents 2.25 (0.70 – 7.25), 0.175 0.35 (0.03 – 4.00), 0.399

Desaturation 3.91 (2.14 – 7.15), < 0.0001 2.17 (0.76 – 6.16), 0.145

Arrhythmia 1.54 (0.18 – 12.91), 0.688 ---

Hypotension 1.77 (1.22 – 2.56), 0.002 0.96 (0.53 – 1.71), 0.882

Need of vasoactive drugs 2.07 (1.42 – 3.02), < 0.0001 0.84 (0.42 – 1.66), 0.608

Postoperative characteristics

No reversal of neuromuscular blockade 1.39 (0.99 – 1.94), 0.051 1.42 (0.85 – 2.38), 0.175

Postoperative residual curarisation 4.17 (1.45 – 12.00), 0.008 3.30 (1.20 – 9.07), 0.021

Odds ratio according to one unit increase of variable; PBW, predicted body weight; PEEP, positive end-expiratory pressure; Peak pressure: peak inspiratory pressure; FiO2, inspired fraction of oxygen; BMI: body mass index; COPD: chronic obstructive pulmonary disease; SpO2: peripheral oxygen saturation; Hb: haemoglobin

134

eTable 12. Univariate and Multivariate Logistic Regression in Patients with ARISCAT ≥ 26 (n = 2144)

Variables Univariate ModelOR (95% CI), p value

Multivariate ModelOR (95% CI), p value

Ventilation parameters

Tidal volume, ml/kg PBW 0.98 (0.91 – 1.05), 0.523 ---

PEEP, cmH2O 1.07 (1.02 – 1.12), 0.003 1.07 (1.01 – 1.14), 0.031

Peak pressure, cmH2O 1.01 (0.99 – 1.03), 0.371 ---

FiO2 1.00 (0.99 – 1.01), 0.863 ---

Characteristics of patients

Male sex 1.09 (0.89 – 1.34), 0.411 ---

BMI, kg/m2 1.01 (0.99 – 1.02), 0.335 ---

COPD 1.30 (0.94 – 1.81), 0.116 1.68 (1.06 – 2.68), 0.028

Age, years ≤ 50 51 – 80 > 80

1 (Reference)1.55 (1.18 – 2.04), 0.0022.38 (1.60 – 3.53), < 0.0001

1 (Reference)1.70 (1.17 – 2.46), 0.0052.81 (1.61 – 4.88), < 0.0001

Functional Status Independent Partially Dependent Totally Dependent

1 (Reference)1.84 (1.36 – 2.50), < 0.00011.70 (0.81 – 3.58), 0.164

1 (Reference)1.21 (0.74 – 1.98), 0.4480.99 (0.32 – 3.12), 0.997

Current Smoker 1.03 (0.80 – 1.34), 0.794 ---

Respiratory Infection < 30 days 1.13 (0.80 – 1.61), 0.474 ---

Preoperative SpO2, % ≥ 96 91 – 95 ≤ 90

1 (Reference)1.13 (0.89 – 1.44), 0.3102.42 (1.53 – 3.83), < 0.0001

1 (Reference)1.13 (0.81 – 1.57), 0.4771.83 (0.93 – 3.59), 0.079

Perioperative anaemia (Hb < 10 g/dl) 1.14 (0.87 – 1.49), 0.346 ---

Characteristics of surgery

Non-Laparoscopic surgery 1.66 (1.29 – 2.14), < 0.0001 0.84 (0.63 – 1.14), 0.268

Moment of Surgery Elective Urgency Emergency

1 (Reference)1.84 (1.34 – 2.54), < 0.00012.15 (1.35 – 3.42), 0.001

1 (Reference)1.76 (1.07 – 2.91), 0.0272.61 (1.12 – 6.08), 0.026

Planned Duration of Surgery, hours ≤ 2 2 – 3 > 3

1 (Reference)1.16 (0.86 – 1.56), 0.3201.52 (1.13 – 2.04), 0.006

1 (Reference)1.14 (0.77 – 1.69), 0.5191.31 (0.78 – 2.19), 0.307

LAS VEGAS study

Chap

ter

5

135

Type of Incision Peripheral Abdominal

1 (Reference)0.91 (0.72 – 1.15), 0.424

---

Intraoperative characteristics

Fluid intake, millilitres < 999 1000 – 1999 2000 – 2999 ≥ 3000

1 (Reference)1.11 (0.73 – 1.67), 0.6281.13 (0.74 – 1.72), 0.5741.59 (1.05 – 2.42), 0.029

1 (Reference)1.62 (1.00 – 2.61), 0.0491.65 (0.94 – 2.90), 0.0802.21 (1.24 – 3.95), 0.007

Epidural anaesthesia 1.43 (1.10 – 1.86), 0.008 1.02 (0.72 – 1.46), 0.894

No use of neuromuscular blocking agents 1.03 (0.58 – 1.84), 0.907 ---

Desaturation 3.03 (2.10 – 4.37), < 0.0001 2.20 (1.27 – 3.79), 0.005

Arrhythmia 2.54 (1.22 – 5.32), 0.013 2.87 (1.05 – 7.86), 0.040

Hypotension 1.78 (1.44 – 2.20), < 0.0001 1.20 (0.84 – 1.73), 0.319

Need of vasoactive drugs 1.92 (1.55 – 2.37), < 0.0001 0.97 (0.61 – 1.55), 0.903

Postoperative characteristics

No reversal of neuromuscular blockade 1.43 (1.15 – 1.77), 0.001 1.11 (0.77 – 1.62), 0.568

Postoperative residual curarisation 4.19 (2.00 – 8.75), < 0.0001 3.91 (1.79 – 8.55), 0.001

Odds ratio according to one unit increase of variable; ARISCAT, The Assess Respiratory Risk in Surgical Patients in Catalonia; PBW, predicted body weight; PEEP, positive end-expiratory pressure; Peak pressure: peak inspiratory pressure; FiO2, inspired fraction of oxygen; BMI: body mass index; COPD: chronic obstructive pulmonary disease; SpO2: peripheral oxygen saturation; Hb: haemoglobin

References1. ARDS Definition Task Force, Ranieri VM, Rubenfeld GD, et al. Acute respiratory distress syndrome: the Berlin

Definition. JAMA 2012, 307(23): 2526-332. Canet J, Gallart L, Gomar C, et al. Prediction of Postoperative Pulmonary Complications in a Population-based

Surgical Cohort. Anesthesiology 2010; 113: 1338-50

Chapter 6

Protective ventilation with lower tidal volumes and high PEEP versus conventional ventilation with high tidal volume and low PEEP in patients under general anesthesia for surgery: A systematic review and individual patient data metaanalysis

Serpa Neto A, Hemmes SNT, Barbas CS, Beiderlinden M, Biehl M, Binnekade JM, Canet C, Fernandez-Bustamante A, Futier E, Gajic O, Hedenstierna G, Hollmann MW, Jaber S, Kozian A, Licker M, Lin WQ, Maslow AD, Memtsoudis SG, Reis Miranda D, Moine P, Ng T, Paparella D, Putensen C, Ranieri M, Scavonetto F, Schilling T, Schmid W, Selmo G, Severgnini P, Sprung J, Sundar S, Talmor D, Treschan T, Unzueta C, Weingarten TN, Wolthuis EK, Wrigge H, Gama de Abreu M, Pelosi P, Schultz MJ, for the PROVE Network investigators.Anesthesiology 2015, May 15 [ePub ahead of print]

138

Abstract

Background. Recent studies show that intraoperative mechanical ventilation using low tidal volumes (VT) can prevent postoperative pulmonary complications (PPC). The aim of this individual patient data metaanalysis is to evaluate the individual associations between VT size and PEEP level, and occurrence of PPC.

Methods. Randomized controlled trials comparing protective ventilation (low VT with or without high levels of PEEP) and conventional ventilation (high VT with low PEEP) in patients undergoing general surgery. The primary outcome was development of PPC. Predefined prognostic factors were tested using multivariate logistic regression.

Results. Fifteen randomized controlled trials were included (2.127 patients). There were 97 cases of PPC in 1.118 patients (8.7%) assigned to protective ventilation and 148 cases in 1.009 patients (14.7%) assigned to conventional ventilation (adjusted relative risk [RR], 0.64; 95% confidence interval [CI], 0.46-0.88; p < 0.01). There were 85 cases of PPC in 957 patients (8.9%) assigned to ventilation with low VT and high PEEP levels and 63 cases in 525 patients (12%) assigned to ventilation with low VT and low PEEP levels (adjusted RR, 0.93; 95% CI, 0.64-1.37; p = 0.72). A dose–response relationship was found between the appearance of PPC and VT size (R2 = 0.39), but not between the appearance of PPC and PEEP level (R2 = 0.08).

Conclusion. This data supports the beneficial effects of ventilation with use of low VT in patients undergoing surgery. Further trials are necessary to define the role of intraoperative higher PEEP to prevent PPC during non-open abdominal surgery.

IPD metaanalysis on protective ventilation

Chap

ter

6

139

Introduction

More than 230 million major surgical procedures are undertaken worldwide each year.1 Postoperative complications after major surgery increase resource use and are an important cause of death.2 Postoperative pulmonary complications (PPC) are suggested to have a strong impact on the morbidity and mortality of patients who need major surgery.2

A systematic review and metaanalysis of investigations in patients receiving ventilation during general anaesthesia for surgery suggests benefit from so–called ‘protective’ ventilator strategies that use low tidal volumes (VT) with or without high positive end–expiratory pressure (PEEP) levels.3 Two randomized controlled trials of intraoperative ventilation, published after this metaanalysis, confirm benefit from the combination of low VT and high PEEP levels.4,5 Another recent trial demonstrates no benefit from high PEEP levels with the use of low VT, but shows use of high PEEP levels to be associated with the appearance of intraoperative hypotension and increased need for vasoactive drugs.6 Contrary, a large retrospective study showed that use of low VT during general anaesthesia for surgery is associated with increased 30–day mortality, and the investigators suggest that this negative effect was due to the use of low PEEP.7

To gain a better understanding of the independent role of VT and PEEP on protective mechanical ventilation during surgery, we performed a systematic review and metaanalysis of individual patient data. We aimed to investigate the individual associations between ventilation settings, including VT size and PEEP level, and the appearance of postoperative pulmonary complications. We hypothesize (a) intraoperative ventilation with low VT to protect against postoperative pulmonary complications, and (b) use of high PEEP to add to the beneficial effects of intraoperative ventilation with low VT.

Materials and methods

The full methodology of this metaanalysis, the predefined protocol and the statistical analysis plan has been published previously and is presented in the Supplementary Appendix.8 Due to the high number of patients from randomized controlled trials, we decided to deviate from the original protocol and chose to exclude observational studies (i.e., we used only individual patient data from the randomized controlled trials).

Search strategyWe identified eligible randomized controlled trials by a blind electronic search by two authors of MEDLINE, Cumulative Index to Nursing and Allied Health Literature (CINAHL), Web of Science, and Cochrane Central Register of Controlled Trials (CENTRAL) up to April 2014. The sensitive search strategy combined the following Medical Subject Headings and Keywords ([protective ventilation OR lower tidal volume OR low tidal volume OR positive end-expiratory pressure OR positive end expiratory pressure OR PEEP]). All reviewed articles and cross–referenced studies from retrieved articles were screened for pertinent information.

140

Selection of studiesRandomized controlled trials eligible for this review compared protective with conventional ventilation in adult patients undergoing general anaesthesia for surgery. Protective ventilation was defined as ventilation using low tidal volume (defined as a tidal volume ≤ 8 ml/kg predicted body weight [PBW]) with or without high levels of PEEP (defined as PEEP ≥ 5 cmH2O) and with or without recruitment manoeuvres. Conventional ventilation was defined as ventilation using high tidal volume (> 8 ml/kg PBW) with or without low levels of PEEP (< 5 cmH2O) and without recruitment manoeuvres. The definition of protective and conventional ventilation was made based on several reports in the literature and according to the previously published protocol.3,4,6,8

Authors independently assessed trial eligibility based on titles, abstracts, full–text reports, and further information from investigators as needed. Corresponding authors of retrieved trials were asked to fill a datasheet with ventilation parameters obtained hourly during the surgical procedure. Data from each trial were checked against reported results, and queries were resolved with the principal investigator. Some of the outcomes in this report may differ slightly from those in published original study reports because we standardized outcome definitions and data analyses.

To identify potential sources of bias, we examined concealment of treatment allocation, blinding of clinical outcome assessments and data analyses, the proportion of patients lost to follow–up, and early stopping prior to enrolment of the target sample. We used the Grading of Recommendations Assessment, Development and Evaluation system to rate the overall quality of the evidence. In this system, randomized clinical trials provide high-quality evidence unless limited by important risk of bias, imprecision, inconsistency, indirectness, or high risk of publication bias.

Outcomes

The predefined primary outcome was development of postoperative pulmonary complications during follow–up (composite of postoperative lung injury, pulmonary infection or barotrauma, as defined by the authors in the original studies). Predefined secondary outcomes included in–hospital mortality; intensive care unit (ICU) length of stay; and hospital length of stay.

Statistical analysisAll patients were analysed in the study group to which they were randomized in the original study (intention-to-treat principle). We used 2–sided t–tests to compare respiratory variables during follow–up and likelihood ratio tests to compare statistical models.

For the primary analysis of development of postoperative pulmonary complications, we calculated relative risks (RRs) and 95% confidence intervals (CIs) using logistic regression. The initial model included age, sex, body mass index, type of surgery, ASA (American Society of Anesthesiology score), type of ventilation, highest PEEP used during surgery, highest plateau pressure achieved during surgery, highest compliance achieved during surgery, and presence of risk factor for postoperative pulmonary complications [defined as shock, pneumonia, blood transfusion and/or sepsis]). Variables with p < 0.2 in the univariate analysis are included in the

IPD metaanalysis on protective ventilation

Chap

ter

6

141

multivariate regression. The final model was developed by dropping each variable in turn from the model and conducting a likelihood-ratio test to compare the full and the nested models. We used a significance level of 0.05 as the cut-off to exclude a variable from the model.

To compare in–hospital time to development of postoperative pulmonary complications and in–hospital time to death for the groups under protective or conventional ventilation, we fitted Cox regression models with the same co–variables. Time–to–event was defined as time from the day of surgery to the event in days. Cox proportional–hazards regression models were used to examine simultaneous effects of multiple covariates on outcomes, censoring a patient’s data at the time of death, hospital discharge, or after 30 days. In all models, the categorical outcome variables were tested for trend with the conventional ventilation group as reference. Kaplan–Meier curves were constructed and log–rank tests were used to determine the univariate significance of the study variables.

A priori subgroup analyses were used to assess the effect of VT in the following predefined subgroups: 1) ASA score (< 3 vs. ≥ 3); 2) presence of risk factors for postoperative pulmonary complications (yes or no, defined as pneumonia, shock, transfusion, and/or sepsis); 3) type of ventilation (volume or pressure controlled); 4) type of surgery (cardiac, abdominal, thoracic, or orthopaedic); 5) body mass index (< 17, 18 – 25, 26 – 30, 31 – 35, or > 35 kg/m2); 6) age (< 65 or ≥ 65 years); and 7) sex (male or female).

To assess the individual effects of PEEP on outcome, all analyses were reassessed post-hoc in patients ventilated with low VT (≤ 8 ml/kg PBW) and stratified between those using low (< 5 cmH2O) or high PEEP levels (≥ 5 cmH2O).4 Also, Kaplan-Meier curves of patients ventilated with PEEP ≥ 5 cmH2O were constructed to compare ventilation with tidal volume ≤ 7 ml/kg PBW vs. 8 – 10 ml/kg PBW vs. > 10 ml/kg PBW. These cut-offs were chosen based on the cut-offs usually used in the literature for low (6 ml/kg PBW) and high tidal volume (10 – 12 ml/kg PBW) and the level between them.4-7 Also, in a post-hoc analysis, we analysed the relationship between four cut-offs of PEEP (0-2, 3-5, 6-8 and ≥ 9 cm H2O, with 0-2 cm H2O as the reference) and tidal volume (3-5, 6-8, 9-11 and ≥ 12 ml/kg PBW with ≥ 12 ml/kg PBW as the reference) with the primary outcome. Finally, in a post-hoc analysis, we analysed recruitment manoeuvres as a dichotomous variable in the regression model, using non-recruitment as reference, and adjusted by the same set of co-variables described above.

PROBIT regression analysis was used to characterize the dose–response relationship between the intra–operative VT size and PEEP level and the probability of postoperative pulmonary complications, while adjusting for the same set of covariates used in the final Cox model. A quadratic term was used in the final model for PEEP and tidal volume. The quadratic term was chosen because we hypothesize that the relationship between PEEP, VT and PPC is curvilinear and the highest-degree term is the second degree. This was confirmed by the inspection of the residuals.

All analyses were conducted with SPSS v.20 (IBM SPSS Statistics for Windows, Version 20.0. Armonk, NY: IBM Corp.) or R v.2.12.0. For all analyses two-sided p values < .05 were considered significant.

142

Results

Search results and collection of individual patient dataThe search identified 21 randomized controlled trials of intraoperative ventilation comparing different VT size and PEEP levels. We were not able to collect data from six trials due to the following reasons: the corresponding author could not provide data of interest or had no longer access to the complete database (n = 3),9-11 or the corresponding author could not be contacted (n = 3).12-14 The total enrolment based on 15 trial trials for which individual patient data could be collected was 2.127 patients (Figure 1 and Table 1).4,6,15-26 In one trial the difference between the two groups was restricted to use of recruitment manoeuvres,25 in one trial use of recruitment manoeuvres and PEEP level,6 and in three trials the VT size.18,22,23 In the other trials, both VT size and PEEP level differed between the two arms of the trial. The methodological quality of included trials was high, with 13 trials using concealed randomization, six trials using blind data analysis, and only three trials having minimal lost to follow–up.

Patient characteristics and ventilator settingsPatient characteristics and ventilator settings are shown in Table 2 and Table 3. Patients receiving protective ventilation were ventilated with higher PEEP levels, respiratory rates, plateau pressure,

Table 1. Characteristics of included trials

ml/kg PBW Trials

Wrigge Zupancich Miranda Schilling Wolthuis Lin Weingarten Sundar Treschan Memtsoudis Unzueta Severgnini Futier Maslow Hemmes

Type of surgery General Cardiac Cardiac Thoracic General Thoracic Abdominal Cardiac Abdominal Spine Thoracic Abdominal Abdominal Thoracic Abdominal

Number of centres 01 01 01 01 01 01 01 01 01 01 01 01 07 01 30

Country Germany Italy Dutch Germany Dutch China USA USA Germany USA Spain Italy France USA Europe/USA

Number of patients Protective arm Conventional arm

2933

2112

2321

7535

2426

5052

2020

7574

5249

1014

4000

2827

200200

1616

455434

Validity Concealed allocation Follow-up, % Blinded analysis

Yes95.4No

NS100No

Yes100Yes

Yes100No

Yes100No

NS100No

Yes100No

Yes98.7Yes

Yes100Yes

Yes100Yes

Yes100No

Yes98.3Yes

Yes100Yes

Yes100No

Yes100Yes

Stopped early No No No No No No No No No No No No No No No

Tidal volume, ml/kg PBW Protective arm Conventional arm

612 – 15

810 – 12

6 – 86 – 8

510

612

5 – 610

610

610

612

612

6 – 86 – 8

79

6 – 810 – 12

510

88

PEEP, cmH2O Protective arm Conventional arm

100

102 – 3

105

0 – 50 – 5

100

3 – 50

120

Scale 55

80

88

100

6 – 80

50

120 – 2

Jadad score 3 3 4 3 3 2 3 4 4 4 3 4 4 3 4

NS: not specified; PBW: predicted body weight; PEEP: positive end-expiratory pressure; USA: United States of America

IPD metaanalysis on protective ventilation

Chap

ter

6

143

Table 1. Characteristics of included trials

ml/kg PBW Trials

Wrigge Zupancich Miranda Schilling Wolthuis Lin Weingarten Sundar Treschan Memtsoudis Unzueta Severgnini Futier Maslow Hemmes

Type of surgery General Cardiac Cardiac Thoracic General Thoracic Abdominal Cardiac Abdominal Spine Thoracic Abdominal Abdominal Thoracic Abdominal

Number of centres 01 01 01 01 01 01 01 01 01 01 01 01 07 01 30

Country Germany Italy Dutch Germany Dutch China USA USA Germany USA Spain Italy France USA Europe/USA

Number of patients Protective arm Conventional arm

2933

2112

2321

7535

2426

5052

2020

7574

5249

1014

4000

2827

200200

1616

455434

Validity Concealed allocation Follow-up, % Blinded analysis

Yes95.4No

NS100No

Yes100Yes

Yes100No

Yes100No

NS100No

Yes100No

Yes98.7Yes

Yes100Yes

Yes100Yes

Yes100No

Yes98.3Yes

Yes100Yes

Yes100No

Yes100Yes

Stopped early No No No No No No No No No No No No No No No

Tidal volume, ml/kg PBW Protective arm Conventional arm

612 – 15

810 – 12

6 – 86 – 8

510

612

5 – 610

610

610

612

612

6 – 86 – 8

79

6 – 810 – 12

510

88

PEEP, cmH2O Protective arm Conventional arm

100

102 – 3

105

0 – 50 – 5

100

3 – 50

120

Scale 55

80

88

100

6 – 80

50

120 – 2

Jadad score 3 3 4 3 3 2 3 4 4 4 3 4 4 3 4

NS: not specified; PBW: predicted body weight; PEEP: positive end-expiratory pressure; USA: United States of America

and higher PaCO2 levels during intraoperative ventilation, as compared to those receiving conventional ventilation. VT was higher in patients who received conventional ventilation during the whole period of ventilation, as compared to patients receiving protective ventilation.

Associations between intraoperative ventilator settings and the primary and secondary endpointsThe appearance of postoperative pulmonary complications was lower in patients receiving protective ventilation compared to patients receiving conventional ventilation (adjusted relative risk [RR], 0.64; 95% confidence interval [CI], 0.46–0.88; p < 0.01) (Table 4 and Figure 2). In–hospital mortality and length of stay in ICU and hospital were similar between the two groups, though patients who developed a PPC had a higher ICU length of stay (6.3 vs. 1.1 days, p < 0.01), a higher hospital length of stay (20.6 vs. 17.1 days, p = 0.011), and died more frequently (6.8 vs. 1.5%, p < 0.01). There was no significant interaction for the effects of protective ventilation on primary outcome according to predefined subgroup analyses, like the ASA score (p = 0.96 for interaction), type of surgery (p = 0.44 for interaction), body mass index (p = 0.77 for interaction) and sex (p = 0.85 for interaction) (Figure 3).

Associations between PEEP levels and the primary and secondary endpoints in patients ventilated with low VT

144

Figure 1. Trial flow

Figure 2. Time to postoperative pulmonary complications, composite endpoint and in-hospital mortality for protective and conventional ventilationCox regression models adjusted for age, ASA, and presence of risk factor for postoperative pulmonary complications. HR: hazard ratio; CI: confidence interval

IPD metaanalysis on protective ventilation

Chap

ter

6

145

Table 5 and 6 presents characteristics and outcome for patients ventilated with low VT and high or low PEEP levels. The appearance of postoperative pulmonary complications was not different for patients receiving high or low PEEP levels in these patients (adjusted RR, 0.93; 95% CI, 0.64–1.37; p = 0.72) (Table 7, Figure 4). In–hospital mortality and length of stay in ICU and hospital were also similar between these two groups. There was no association between higher cut-offs of PEEP and the incidence of PPC compared to 0-2 cmH2O of PEEP (Figure 5). There was no significant interaction for the effects of PEEP on primary outcome according to predefined subgroup analyses (Figure 6). Also, the appearance of postoperative pulmonary complications was not different for patients receiving recruitment manoeuvres (adjusted RR for the whole cohort, 0.72; 95% CI, 0.49–1.05; p = 0.09 and adjusted RR for patients ventilated with tidal volume ≤ 8 ml/kg PBW, 0.84; 95% CI, 0.54–1.29; p = 0.84).

Associations between tidal volume size and the primary and secondary endpoints in patients ventilated with high PEEPIn patients ventilated with PEEP ≥ 5 cmH2O, the appearance of postoperative pulmonary complications was lower only in patients receiving tidal volume ≤ 7 ml/kg PBW compared to patients ventilated with tidal volume > 10 ml/kg PBW (adjusted RR, 0.40; 95% CI, 0.21–0.78;

Figure 3. Relative risk for Study Outcomes According to Subgroups (Protective vs. Conventional Ventilation)The size of the squares is proportional to the number of patients in the subgroup. ASA: American Society of Anesthesiologists; CI: confidence interval

146

p < 0.01) (Figure 7). Compared to tidal volume ≥ 12 ml/kg PBW, patients ventilated with tidal volume between 6-8 and 3-5 ml/kg PBW presented a lower incidence of PPC (Figure 8). In–hospital mortality was similar between the groups. There was no significant interaction for the effects of tidal volume on primary outcome according to predefined subgroup analyses (Figure 9).

Dose–response relationship between PEEP level and tidal volume size and postoperative pulmonary complicationsDose–response relationship curves between intraoperative tidal volume size and PEEP levels and appearance of postoperative pulmonary complications are shown in Figure 10A and 10B. A dose–response relationship was found between the appearance of PPC and VT size (R2 for mean quadratic term = 0.39), but not between the appearance of PPC and PEEP level (R2 = 0.08).

Figure 4. Time to postoperative pulmonary complications, composite endpoint and in-hospital mortality for patients ventilated with low tidal volumes and high or low levels of PEEPCox regression models adjusted for age, ASA, and presence of risk factor for postoperative pulmonary complications. HR: hazard ratio; CI: confidence interval; PEEP: positive end-expiratory pressure

Figure 5. Relative risk of postoperative pulmonary complications according to different levels of PEEP and using 0 – 2 cmH2O as reference

IPD metaanalysis on protective ventilation

Chap

ter

6

147

Table 2. Baseline characteristics of included patients

Characteristics Protective Ventilation (n = 1,118)

Conventional Ventilation (n = 1,009)

Age, years 63.2 ± 12.8 64.7 ± 11.9

Female, No. (%) 423 (38) 383 (38)

Body mass index, kg/m2 25.7 ± 4.4 25.7 ± 4.4

ASA, No. (%) Median (IQR) 1 2 3 4

2.0 (2 – 3)110 (10)557 (50)429 (38)22 (2)

2.0 (2 – 3)109 (11)500 (50)379 (37)21 (2)

Type of surgery, No. (%) Cardiac Thoracic Abdominal Spine

119 (11)196 (17)793 (71)10 (1)

107 (11)119 (12)769 (76)14 (1)

Risk factor for PPC, No. (%)a

Yes Pneumonia Sepsis Transfusion Shock

143 (13)5 (0.5)5 (0.5)89 (8)44 (4)

149 (15)10 (1)10 (1)89 (9)40 (4)

ASA: American Society of Anesthesiologists; IQR: interquartile range; PPC: postoperative pulmonary complicationsaIndividual patients could have more than one risk factor

Discussion

This individual patient metaanalysis of 2.127 patients ventilated under general anesthesia for surgery from 15 randomized controlled trials shows that intraoperative protective ventilation protects the lung from postoperative pulmonary complications. We found that intraoperative low VT was associated with reduced PPC.

In the intensive care unit, following the publication of ARDSNet low–VT trial in patients with the Acute Respiratory Distress Syndrome (ARDS),27 there has been a progressive decrease in VT size over the last decade from more than 12 ml/kg to less than 9 ml/kg.28-30 These changes were supported by numerous preclinical studies in animals showing that ventilation with high VT was associated with lung inflammation and injury,31 worse oxygenation,32 and vascular dysfunction,33 even in healthy lungs. In the operating room VT size remained unchanged, despite numerous randomized controlled trials suggesting benefit of low VT during intraoperative ventilation.34,35 Lack of knowledge of the existence and under–recognition of postoperative pulmonary complications, as well as the idea that shorter duration of intraoperative ventilation may be less injurious than longer duration of ventilation in the intensive care unit may explain the absence of ventilation

148

practice changes in the operating room.2-4 The present analysis is in accordance with the findings of a previous systematic review and metaanalysis,3 and three randomized controlled trials and showing the benefits of protective ventilation during general anesthesia for surgery.4-6 This metaanalysis helps further in the interpretation and understanding of the individual effects of tidal volume and PEEP.

Experimental studies suggest that high PEEP levels minimize cyclical alveolar collapse and corresponding shear injury to the lungs in patients with ARDS.36,37 Based on this observation, it has been suggested that high PEEP levels could benefit patients with ARDS.38 Randomized controlled trials comparing high PEEP levels with low PEEP levels and one metaanalysis, however, suggest only benefit of high PEEP levels in patients who suffered from severe ARDS.38 Ventilation strategies that use high PEEP levels are associated with potentially dangerous side–effects, including hemodynamic depression and lung overdistention, which could further outweigh the potential benefits.39,40 This was also found in the last randomized controlled trial comparing high with low PEEP levels in patients under intraoperative ventilation with low VT.6 The results of

Figure 6. Relative risk for Study Outcomes According to Subgroups (High vs. Low PEEP)The size of the squares is proportional to the number of patients in the subgroup. ASA: American Society of Anesthesiologists; CI: confidence interval

IPD metaanalysis on protective ventilation

Chap

ter

6

149

Tabl

e 3.

Res

pira

tory

var

iabl

es d

urin

g su

rger

y

Varia

ble

Begi

nnin

g of

Pro

cedu

reM

iddl

e of

Pro

cedu

reEn

d of

Pro

cedu

re

Prot

ectiv

eCo

nven

tiona

lp

valu

ePr

otec

tive

Conv

entio

nal

p va

lue

Prot

ectiv

eCo

nven

tiona

lp

valu

e

Tida

l vol

ume,

ml/k

g PB

W7.

3 ±

1.0

[1,1

14]

10.8

± 1

.5[9

18]

< 0.

017.

8 ±

1.3

[739

]10

.0 ±

1.9

[671

]<

0.01

7.1

± 1.

1[1

,015

]10

.3 ±

1.2

[901

]<

0.01

Plat

eau

pres

sure

, cm

H2O

18.8

± 5

.9[9

50]

15.9

± 4

.8[8

25]

< 0.

0121

.3 ±

6.0

[527

]16

.5 ±

5.1

[466

]<

0.01

18.4

± 5

.4[7

56]

16.8

± 4

.8[6

40]

< 0.

01

PEEP

, cm

H2O

8.6

± 3.

4[1

,011

]1.

3 ±

1.8

[911

]<

0.01

7.3

± 5.

0[7

23]

1.1

± 1.

6[6

20]

< 0.

016.

0 ±

4.6

[1,0

86]

1.1

± 1.

9[9

77]

< 0.

01

Resp

irato

ry ra

te,

mpm

12.4

± 2

.8[9

46]

9.9

± 2.

2[8

36]

< 0.

0113

.0 ±

3.5

[569

]10

.3 ±

2.4

[473

]<

0.01

15.1

± 5

.6[7

96]

10.3

± 2

.8[7

15]

< 0.

01

PaO

2 / F

iO2,

mm

Hg40

4.4

± 14

8.0

[321

]41

5.2

± 16

0.3

[233

]0.

4116

9.1

± 19

4.1

[249

]19

7.9

± 22

3.7

[203

]0.

1433

0.0

± 14

8.5

[371

]30

3.7

± 13

5.9

[281

]0.

02

PaCO

2, m

mHg

42.4

± 6

.0[3

21]

38.5

± 7

.1[2

33]

< 0.

0143

.5 ±

6.8

[249

]38

.7 ±

8.0

[203

]<

0.01

43.7

± 7

.9[3

71]

39.1

± 6

.3[2

81]

< 0.

01

Arte

rial p

H7.

39 ±

0.0

6[3

21]

7.41

± 0

.05

[233

]<

0.01

7.34

± 0

.06

[249

]7.

37 ±

0.0

6[2

03]

< 0.

017.

33 ±

0.0

8[3

71]

7.34

± 0

.10

[281

]0.

17

MPM

: mov

emen

ts p

er m

inut

e; P

BW: p

redi

cted

bod

y w

eigh

t; PE

EP: p

ositi

ve e

nd-e

xpira

tory

pre

ssur

e

150

Table 4. Clinical outcomes in patients undergoing general anesthesia for surgery

OutcomesProtective Ventilation(n = 1,118)

Conventional Ventilation(n = 1,009)

Adjusted RR (95% CI)a p value

Postoperative Pulmonary Complications Acute respiratory distress syndrome Barotrauma Suspected pulmonary infection

97 (8.7)20 (1.8)12 (1.1)79 (7.1)

148 (14.7)51 (5.1)29 (2.9)101 (10.0)

0.64 (0.46 – 0.88)0.45 (0.24 – 0.83)0.39 (0.17 – 0.92)0.83 (0.58 – 1.20)

< 0.010.010.030.33

In-Hospital Mortality 22 (2.0) 20 (2.1) 1.17 (0.52 – 2.62) 0.70

Length of ICU stay, days 1 (0 – 2) 1 (0 – 2) –0.20 (–1.41 to 1.00)b 0.73

Length of hospital stay, days 10 (7 – 18) 11 (7 – 18) –0.61 (–2.80 to 1.57)b 0.58

CI: confidence interval; ICU: intensive care unit; RR: relative riska Multivariate regression with the outcome of interest as dependent variable; Ventilation group, age, ASA, and presence of risk factor as independent variablesb Coefficient from a corresponding linear regression model using the same independent variables and random effect as the above-described model

Table 5. Baseline characteristics of included patients ventilated with low tidal volumes

Characteristics High PEEP (n = 957) Low PEEP (n = 525)

Age, years 63.6 ± 12.8 64.2 ± 12.8

Female, No. (%) 350 (37) 200 (38)

Body mass index, kg/m2 25.9 ± 4.4 25.1 ± 4.3

ASA, No. (%) Median (IQR) 1 2 3 4

2.0 (2 – 3)86 (9)488 (51)344 (36)29 (3)

2.0 (2 – 3)63 (12)241 (46)205 (39)16 (3)

Type of surgery, No. (%) Cardiac Thoracic Abdominal Spine

139 (14)70 (8)738 (77)10 (1)

77 (15)53 (10)395 (75)0 (0)

Risk factor for PPC, No. (%)a

Yes Pneumonia Sepsis Transfusion Shock

124 (13)10 (1)5 (0.5) 71 (7)38 (4)

37 (7)10 (2)3 (0.5)19 (4)5 (1)

ASA: American Society of Anesthesiology; PEEP: positive end-expiratory pressure; PPC: postoperative pulmonary complication; aIndividual patients could have more than one risk factor

IPD metaanalysis on protective ventilation

Chap

ter

6

151

Tabl

e 6.

Res

pira

tory

var

iabl

es d

urin

g su

rger

y in

pati

ents

ven

tilat

ed w

ith lo

w ti

dal v

olum

es

Varia

ble

Begi

nnin

g of

Pro

cedu

reM

iddl

e of

Pro

cedu

reEn

d of

Pro

cedu

re

Hig

h PE

EPLo

w P

EEP

p va

lue*

Hig

h PE

EPLo

w P

EEP

p va

lue*

Hig

h PE

EPLo

w P

EEP

p va

lue*

Tida

l Vol

ume,

ml/k

g PB

W7.

5 ±

1.0

[827

]7.

8 ±

0.8

[484

]0.

127.

8 ±

0.9

[406

]7.

8 ±

0.9

[376

]0.

956.

7 ±

0.9

[526

]6.

9 ±

1.0

[345

]0.

11

Plat

eau

pres

sure

, cm

H 2O

19.0

± 5

.7[8

16]

16.0

± 4

.5[4

62]

< 0.

0121

.1 ±

6.0

[426

]17

.3 ±

5.5

[358

]<

0.01

18.4

± 5

.5[6

37]

16.7

± 4

.3[3

29]

< 0.

01

PEEP

, cm

H 2O

8.8

± 3.

3[9

04]

1.2

± 1.

2[4

62]

< 0.

017.

7 ±

5.0

[626

]1.

1 ±

1.3

[455

]<

0.01

6.6

± 4.

5[9

45]

1.0

± 1.

4[5

25]

< 0.

01

Resp

irato

ry ra

te, b

pm12

.4 ±

2.8

[811

]11

.4 ±

2.1

[460

]<

0.01

12.9

± 3

.6[4

68]

11.8

± 2

.5[3

59]

< 0.

0115

.6 ±

5.9

[681

]12

.0 ±

2.9

[339

]<

0.01

PaO

2 / F

iO2,

mm

Hg42

2.8

± 14

5.7

[249

]34

2.8

± 14

0.5

[73]

< 0.

0117

4.2

± 22

0.2

[180

]14

8.8

± 10

3.0

[76]

0.33

319.

3 ±

164.

6[2

78]

360.

7 ±

127.

9[1

34]

0.01

PaCO

2, m

mHg

42.2

± 5

.8[2

49]

43.5

± 6

.9[7

3]0.

1044

.0 ±

7.1

[180

]42

.6 ±

6.5

[76]

0.17

43.7

± 8

.3[2

78]

43.0

± 6

.0[1

34]

0.38

Arte

rial p

H7.

39 ±

0.0

6[2

49]

7.39

± 0

.07

[73]

0.79

7.34

± 0

.06

[180

]7.

34 ±

0.0

6[7

6]0.

867.

34 ±

0.0

6[2

78]

7.33

± 0

.10

[134

]0.

19

MPM

: bre

aths

per

min

ute;

PBW

: pre

dict

ed b

ody

wei

ght;

PEEP

: pos

itive

end

-exp

irato

ry p

ress

ure;

*Hi

gher

PEE

P vs

Low

er P

EEP

152

this metaanalysis suggest no benefit from high PEEP levels with use of low VT. Thus, high PEEP should not be standard practice, despite the suggestions of an earlier observational study.7

Recently, a large and well–powered randomized controlled trial in France4 confirmed the beneficial effects of protective ventilation in intermediate-risk and high-risk patients undergoing major surgery. However, protection in this trial could have come from low VT, intermediate levels of PEEP, recruitment manoeuvres or from the combination of the three. Indeed, the use of high tidal volume in the conventional arm could be associated with more harm than beneficial of low tidal volume in protective arm. In an attempt to understand the individual effect of PEEP, an international randomized controlled trial evaluated the effects of high PEEP levels with use of low VT.6 High PEEP levels did not prevent postoperative pulmonary complications, but was associated with more hemodynamic compromise.6

Figure 7. Time to postoperative pulmonary complications, composite endpoint and in-hospital mortality for patients ventilated with PEEP ≥ 5 cmH2O and tidal volume ≤ 7 vs. 8 – 10 vs. > 10 ml/kg PBWCox regression models adjusted for age, ASA, and presence of risk factor for postoperative pulmonary complications. HR: hazard ratio; CI: confidence interval; PBW: predicted body weight

Figure 8. Relative risk of postoperative pulmonary complications according to different tidal volumes and using ≥ 12 ml/kg PBW of tidal volume as referencePBW: predicted body weight

IPD metaanalysis on protective ventilation

Chap

ter

6

153

The absence of an association between a protective ventilation strategy and a lower mortality rate could be expected, since mortality of surgical patients is very low in general, and only 1.2% in the cohort of patients included in the present analysis. However, while we did no found differences in mortality and hospital length of stay in in the different ventilation groups, patients who developed a PPC had a higher ICU length of stay, a higher hospital length of stay, and died more frequently.

In this metaanalysis, variability in treatment over time was overcome by conducting a pooled analysis of data on individual patients. The use of these data allowed us to update the number of patients and follow–up after the original published reports. With individual patient data we have enough power to study different subgroups and also to assess the individual effects of PEEP and tidal volume. Also, to date this study included data on the largest population available for comparison of the benefits of protective ventilation in the surgical setting and postoperative outcome.41

Figure 9. Relative risk for Study Outcomes According to Subgroups (≤ 7 ml/kg PBW vs. > 10 ml/kg PBW) The size of the squares is proportional to the number of patients in the subgroup. ASA: American Society of Anesthesiologists; CI: confidence interval; PBW: predicted body weight

154

This metaanalysis knows limitations. First, not all investigators could provide the data, and, therefore, data from six identified studies were not included.9-14 However, the results of a classical metaanalysis including all but one study14 are in agreement with those found in the present analysis. Thus the assumption can be made that the included studies are reliable representatives of all studies of protective ventilation during surgery.5 Second, since the diagnosis of postoperative lung injury is based on clinical criteria, misclassification of patients might underestimate the observed effect, but this factor should have equally affect the different groups analysed. Third, we do not have information on some important factors that could contribute to the development of postoperative complications, including but not limited to

Figure 10. PROBIT logistic regression showing the dose-relationship curve between the A) mean tidal volume (ml/kg PBW) and B) mean PEEP (cmH2O) used in surgery and the probability of postoperative pulmonary complications Solid line: mean quadratic term; Dashed line: 95% confidence interval. The line represents the quadratic term fitting all the points. The flat line in the PEEP graph suggests that there is neither a positive nor a negative association between a higher level of PEEP and the development of postoperative pulmonary complications. PBW: predicted body weight; PEEP: positive end-expiratory pressure

IPD metaanalysis on protective ventilation

Chap

ter

6

155

fluid balance, use of colloids, recruitment manoeuvres and postoperative analgesia. Fourth, since we collected sufficient data on other PPCs, we deviate from the primary outcome stated in the preliminary protocol (‘development of ARDS’)8 to a stronger outcome (‘development of any PPC’), since PPCs were reported in the majority of retrieved studies. Fifth, different types of surgery were analysed and can be a confounding factor. However, no interaction was found between type of surgery and primary outcome according to the predefined subgroup analyses. Finally, due to the variability between the effects on primary outcome, our analysis on PEEP could be underpowered. In fact the highest PEEP quartile was lower than 1 compared to 0-2 cm H2O PEEP. However, the moderate PEEP group 6-8 cmH2O showed a non-significant increase, and not decrease, in the risk of PPC. Higher PEEP was found not effective to reduce PPC when protective tidal volumes were used during open abdominal surgery.6 Also, most of the studies included in the analysis were not a priori conducted to evaluate PEEP effects. Additional studies are required to test the hypothesis that high levels of PEEP during different type of surgery can protect our patients from postoperative respiratory complications.

In conclusion, this individual patient data metaanalysis shows that intraoperative ventilation with low VT protects against postoperative pulmonary complications. Further trials are necessary to define the role of intraoperative higher PEEP to prevent PPC during non-open abdominal surgery.

Financial supportSupport was provided solely from institutional and/or departmental sources.

Table 7. Clinical outcomes in patients undergoing general anesthesia for surgery ventilated with lower tidal volumes

Outcomes High PEEP(n = 957)

Low PEEP(n = 525) Adjusted RR (95% CI)a p value

Postoperative Pulmonary Complications Acute respiratory distress syndrome Barotrauma Suspected pulmonary infection

85 (8.9)20 (2.1)12 (1.3)66 (6.9)

63 (12)15 (2.8)9 (1.8)55 (10.4)

0.93 (0.64 – 1.37)0.82 (0.38 – 1.74)0.66 (0.25 – 1.77)0.81 (0.54 – 1.23)

0.720.600.410.33

In-Hospital Mortality 18 (1.9) 7 (1.3) 1.34 (0.47 – 3.78) 0.57

Length of ICU stay, days 0 (0 – 1) 1 (1 – 2) –0.31 (–1.91 to 1.27)b 0.69

Length of hospital stay, days 10 (7 – 18) 11 (8 – 18) –0.48 (–3.04 to 2.07)b 0.71

CI: confidence interval; ICU: intensive care unit; PEEP: positive end expiratory pressure; RR: relative riskaMultivariate regression with the outcome of interest as dependent variable; Ventilation group, age, ASA, and presence of risk factor as independent variablesbCoefficient from a corresponding linear regression model using the same independent variables and random effect as the above-described model

156

References1. Pearse RM, Moreno RP, Bauer P, Pelosi P, Metnitz P, Spies C, Vallet B, Vincent JL, Hoeft A, Rhodes A; European Surgical

Outcomes Study (EuSOS) group for the Trials groups of the European Society of Intensive Care Medicine and the European Society of Anaesthesiology: Mortality after surgery in Europe: A 7 day cohort study. Lancet 2012; 380:1059-65

2. Canet J, Gallart L, Gomar C, Paluzie G, Vallès J, Castillo J, Sabate S, Mazo V, Briones Z, Sanchis J; ARISCAT Group: Prediction of postoperative pulmonary complications in a population-based surgical cohort. Anesthesiology 2010; 113:1338-50

3. Serpa Neto A, Cardoso SO, Manetta JA, Pereira VG, Esposito DC, Pasqualucci Mde O, Damasceno MC, Schultz MJ: Association between use of lung-protective ventilation with lower tidal volumes and clinical outcomes among patients without acute respiratory distress syndrome: a metaanalysis. JAMA 2012; 308:1651-9

4. Futier E, Constantin JM, Paugam-Burtz C, Pascal J, Eurin M, Neuschwander A, Marret E, Beaussier M, Gutton C, Lefrant JY, Allaouchiche B, Verzilli D, Leone M, De Jong A, Bazin JE, Pereira B, Jaber S; IMPROVE Study Group: A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. NEJM 2013; 369:428-37

5. Severgnini P, Selmo G, Lanza C, Chiesa A, Frigerio A, Bacuzzi A, Dionigi G, Novario R, Gregoretti C, de Abreu MG, Schultz MJ, Jaber S, Futier E, Chiaranda M, Pelosi P: Protective mechanical ventilation during general anesthesia for open abdominal surgery improves postoperative pulmonary function. Anesthesiology 2013; 118:1307-21

6. The PROVE Network Investigators: Higher versus lower positive end-expiratory pressure during general anaesthesia for open abdominal surgery - The PROVHILO trial. Lancet 2014; 384:495-503

7. Levin MA, McCormick PJ, Lin HM, Hosseinian L, Fischer GW: Low intraoperative tidal volume ventilation with minimal PEEP is associated with increased mortality. Br J Anaesth 2014; 113:97-108

8. Serpa Neto A, Hemmes SNT, Gama de Abreu M, Pelosi P, Schultz MJ; Protective Ventilation Network (PROVENet): Protocol for a systematic review and individual patient metaanalysis of benefit of so-called lung-protective ventilation-settings in patients under general anesthesia for surgery. Syst Rev 2014; 3:2-6

9. Chaney MA, Nikolov MP, Blakeman BP, Bakhos M: Protective ventilation attenuates postoperative pulmonary dysfunction in patients undergoing cardiopulmonary bypass. J Cardiothorac Vasc Anesth 2000; 14:514-8

10. Koner O, Celebi S, Balci H, Cetin G, Karaoglu K, Cakar N: Effects of protective and conventional mechanical ventilation on pulmonary function and systemic cytokine release after cardiopulmonary bypass. Intensive Care Med 2004; 30:620-6

11. Michelet P, D’Journo XB, Roch A, Doddoli C, Marin V, Papazian L, Decamps I, Bregeon F, Thomas P, Auffray JP: Protective ventilation influences systemic inflammation after esophagectomy: A randomized controlled study. Anesthesiology 2006; 105:911-9

12. Cai H, Gong H, Zhang L, Wang Y, Tian Y: Effect of low tidal volume ventilation on atelectasis in patients during general anesthesia: A computed tomographic scan. J Clin Anesth 2007; 19:125-9

13. Yang M, Ahn HJ, Kim K, Kim JA, Yi CA, Kim MJ, Kim HJ: Does a protective ventilation strategy reduce the risk of pulmonary complications after lung cancer surgery?: A randomized controlled trial. Chest 2011; 139:530-7

14. Ahn HJ, Kim JA, Yang M, Shim WS, Park KJ, Lee JJ: Comparison between conventional and protective one-lung ventilation for ventilator-assisted thoracic surgery. Anaesth Intensive Care 2012; 40:780-8

15. Wrigge H, Uhlig U, Zinserling J, Behrends-Callsen E, Ottersbach G, Fischer M, Uhlig S, Putensen C: The effects of different ventilatory settings on pulmonary and systemic inflammatory responses during major surgery. Anesth Analg 2004; 98:775-81

16. Zupancich E, Paparella D, Turani F, Munch C, Rossi A, Massaccesi S, Ranieri VM: Mechanical ventilation affects inflammatory mediators in patients undergoing cardiopulmonary bypass for cardiac surgery: A randomized clinical trial. J Thorac Cardiovasc Surg 2005; 130:378-83

17. Reis Miranda D, Gommers D, Struijs A, Dekker R, Mekel J, Feelders R, Lachmann B, Bogers AJ: Ventilation according to the open lung concept attenuates pulmonary inflammatory response in cardiac surgery. Eur J Cardiothorac Surg 2005; 28:889-95

18. Schilling T, Kozian A, Huth C, Bühling F, Kretzschmar M, Welte T, Hachenberg T: The pulmonary immune effects of mechanical ventilation in patients undergoing thoracic surgery. Anesth Analg 2005; 101:957-65

19. Wolthuis EK, Choi G, Dessing MC, Bresser P, Lutter R, Dzoljic M, van der Poll T, Vroom MB, Hollmann M, Schultz MJ: Mechanical ventilation with lower tidal volumes and positive end-expiratory pressure prevents pulmonary inflammation in patients without preexisting lung injury. Anesthesiology 2008; 108:46-54

20. Lin WQ, Lu XY, Cao LH, Wen LL, Bai XH, Zhong ZJ: Effects of the lung protective ventilatory strategy on proinflammatory cytokine release during one-lung ventilation. Ai Zheng 2008; 27:870-3

21. Weingarten TN, Whalen FX, Warner DO, Gajic O, Schears GJ, Snyder MR, Schroeder DR, Sprung J: Comparison of two ventilatory strategies in elderly patients undergoing major abdominal surgery. Br J Anaesth 2010; 104:16-22

22. Sundar S, Novack V, Jervis K, Bender SP, Lerner A, Panzica P, Mahmood F, Malhotra A, Talmor D: Influence of low tidal volume ventilation on time to extubation in cardiac surgical patients. Anesthesiology 2011; 114:1102–10

23. Treschan TA, Kaisers W, Schaefer MS, Bastin B, Schmalz U, Wania V, Eisenberger CF, Saleh A, Weiss M, Schmitz A,

IPD metaanalysis on protective ventilation

Chap

ter

6

157

Kienbaum P, Sessler DI, Pannen B, Beiderlinden M: Ventilation with low tidal volumes during upper abdominal surgery does not improve postoperative lung function. Br J Anaesth 2012; 109:263-71

24. Memtsoudis SG, Bombardieri AM, Ma Y, Girardi FP: The effect of low versus high tidal volume ventilation on inflammatory markers in healthy individuals undergoing posterior spine fusion in the prone position: A randomized controlled trial. J Clin Anesth 2012; 24:263-9

25. Unzueta C, Tusman G, Suarez-Sipmann F, Böhm S, Moral V: Alveolar recruitment improves ventilation during thoracic surgery: A randomized controlled trial. Br J Anaesth 2012; 108:517-24

26. Maslow AD, Stafford TS, Davignon KR, Ng T: A randomized comparison of different ventilator strategies during thoracotomy for pulmonary resection. J Thorac Cardiovasc Surg 2013;146:38-44

27. Acute Respiratory Distress Syndrome Network: Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. NEJM 2000; 342:1301-8

28. Azevedo LC, Park M, Salluh JI, Rea-Neto A, Souza-Dantas VC, Varaschin P, Oliveira MC, Tierno PF, Dal-Pizzol F, Silva UV, Knibel M, Nassar AP Jr, Alves RA, Ferreira JC, Teixeira C, Rezende V, Martinez A, Luciano PM, Schettino G, Soares M; The ERICC (Epidemiology of Respiratory Insufficiency in Critical Care) investigators: Clinical outcomes of patients requiring ventilatory support in Brazilian intensive care units: A multicenter, prospective, cohort study. Crit Care 2013; 17:R63

29. Esteban A, Frutos-Vivar F, Muriel A, Ferguson ND, Penuelas O, Abraira V, Raymondos K, Rios F, Nin N, Apezteguía C, Violi DA, Thille AW, Brochard L, González M, Villagomez AJ, Hurtado J, Davies AR, Du B, Maggiore SM, Pelosi P, Soto L, Tomicic V, D’Empaire G, Matamis D, Abroug F, Moreno RP, Soares MA, Arabi Y, Sandi F, Jibaja M, Amin P, Koh Y, Kuiper MA, Bülow HH, Zeggwagh AA, Anzueto A: Evolution of Mortality over Time in Patients Receiving Mechanical Ventilation. Am J Respir Crit Care Med 2013; 188:220-30

30. Wolthuis EK, Vlaar AP, Choi G, Roelofs JJ, Juffermans NP, Schultz MJ: Mechanical ventilation using non-injurious ventilation settings causes lung injury in the absence of pre-existing lung injury in healthy mice. Crit Care 2009; 13:R1

31. Hegeman MA, Hemmes SN, Kuipers MT, Bos LD, Jongsma G, Roelofs JJ, van der Sluijs KF, Juffermans NP, Vroom MB, Schultz MJ: The extent of ventilator-induced lung injury in mice partly depends on duration of mechanical ventilation. Crit Care Res Pract 2013; 2013:435236

32. Menendez C, Martinez-Caro L, Moreno L, Nin N, Moral-Sanz J, Morales D, Cogolludo A, Esteban A, Lorente JA, Perez-Vizcaino F: Pulmonary vascular dysfunction induced by high tidal volume mechanical ventilation. Crit Care Med 2013; 41:e149-55

33. Gajic O, Dara SI, Mendez JL, Adesanya AO, Festic E, Caples SM, Rana R, St Sauver JL, Lymp JF, Afessa B, Hubmayr RD: Ventilator-associated lung injury in patients without acute lung injury at the onset of mechanical ventilation. Crit Care Med 2004; 32:1817-24

34. Jaber S, Coisel Y, Chanques G, Futier E, Constantin JM, Michelet P, Beaussier M, Lefrant JY, Allaouchiche B, Capdevila X, Marret E: A multicentre observational study of intra-operative ventilatory management during general anaesthesia: Tidal volumes and relation to body weight. Anaesthesia 2012; 67:999-1008

35. Fernández-Perez ER, Sprung J, Afessa B, Warner DO, Vachon CM, Schroeder DR, Brown DR, Hubmayr RD, Gajic O: Intraoperative ventilator settings and acute lung injury after elective surgery: A nested case control study. Thorax 2009; 64:121-7

36. Gattinoni L, Caironi P, Cressoni M, Chiumello D, Ranieri VM, Quintel M, Russo S, Patroniti N, Cornejo R, Bugedo G: Lung recruitment in patients with the acute respiratory distress syndrome. NEJM 2006; 354:1775-1786

37. Muscedere JG, Mullen JB, Gan K, Slutsky AS: Tidal ventilation at low airway pressures can augment lung injury. Am J Respir Crit Care Med 1994; 149:1327-34

38. Briel M, Meade M, Mercat A, Brower RG, Talmor D, Walter SD, Slutsky AS, Pullenayegum E, Zhou Q, Cook D, Brochard L, Richard JC, Lamontagne F, Bhatnagar N, Stewart TE, Guyatt G: Higher vs Lower Positive End-Expiratory Pressure in Patients With Acute Lung Injury and Acute Respiratory Distress Syndrome - Systematic Review and Metaanalysis. JAMA 2010; 303:865-73

39. Pinsky MR: The hemodynamic consequences of mechanical ventilation: an evolving story. Intensive Care Med 1997; 23:493-503

40. Wakabayashi K, Wilson MR, Tatham KC, O’Dea KP, Takata M: Volutrauma, but not Atelectrauma, Induces Systemic Cytokine Production by Lung-Marginated Monocytes. Crit Care Med 2014; 42:e49-57

41. Melo MF, Eikermann M: Protect the lungs during abdominal surgery: it may change the postoperative outcome. Anesthesiology 2013; 118:1254-7

Chapter 7

Rationale and study design of PROVHILO – a worldwide multicenter randomized controlled trial on protective ventilation during general anesthesia for open abdominal surgery

Hemmes SNT, Severgnini P, Jaber S, Canet J, Wrigge H, Hiesmayr M, Tschernko EM, Hollmann MW, Binnekade JM, Hedenstierna G, Putensen C, de Abreu MG, Pelosi P, Schultz MJ Trials 2011; 12: 111

160

Abstract

Background. Postoperative pulmonary complications add to the morbidity and mortality of surgical patients, in particular after general anesthesia > 2 hours for abdominal surgery. Whether a protective mechanical ventilation strategy with higher levels of positive end–expiratory pressure (PEEP) and repeated recruitment manoeuvres; the “open lung strategy”, protects against postoperative pulmonary complications is uncertain. The present study aims at comparing a protective mechanical ventilation strategy with a conventional mechanical ventilation strategy during general anesthesia for abdominal non–laparoscopic surgery.

Methods and design. The PROtective Ventilation using HIgh versus LOw positive end–expiratory pressure (“PROVHILO”) trial is a worldwide investigator-initiated multicenter randomized controlled two-arm study. Nine hundred patients scheduled for non–laparoscopic abdominal surgery at high or intermediate risk for postoperative pulmonary complications are randomized to mechanical ventilation with the level of PEEP at 12 cmH2O with recruitment manoeuvres (the lung–protective strategy) or mechanical ventilation with the level of PEEP at maximum 2 cmH2O without recruitment manoeuvres (the conventional strategy). The primary endpoint is any postoperative pulmonary complication.

Discussion. The PROVHILO trial is the first randomized controlled trial powered to investigate whether an open lung mechanical ventilation strategy in short–term mechanical ventilation prevents against postoperative pulmonary complications.

Trial registration. Current Controlled Trials ISRCTN70332574

PROVHILO study design

Chap

ter

7

161

Background

Mechanical ventilation is a life–saving strategy in patients with respiratory failure. There is unequivocal evidence that mechanical ventilation in critically ill patients has the potential to aggravate or even initiate lung injury.1,2 Patients with acute lung injury (ALI) could benefit from measures that prevent repeated collapse and re–expansion of alveoli, including the so–called open lung mechanical ventilation strategy with the use of higher levels of positive end–expiratory pressure (PEEP) and recruitment maneuvers.3 Metaanalysis suggest this approach can waive the need for rescue therapies due to life–threatening hypoxemia,1 and even reduce mortality in patients with more severe ALI.4

Mechanical ventilation is frequently mandatory in patients who undergo surgery. The effects of short term intraoperative mechanical ventilation on pulmonary integrity are less well defined.5 In addition, it is uncertain whether ventilation strategies that use higher levels of PEEP and recruitment manoeuvres during the intraoperative period are beneficial in these patients.6,7 However, higher levels of PEEP could reduce intraoperative atelectasis, decreasing repetitive collapse and re–expansion of dependent lung parts, and thereby attenuating pulmonary inflammation and coagulation.8,9 Use of recruitment manoeuvres to open the lungs has been found to improve the effectiveness of PEEP with regard to gas exchange during general anesthesia.10 Intraoperative use of PEEP does not represent a common practice. Indeed, an observational study conducted in 28 centres in France revealed that most patients undergoing general surgery were ventilated without PEEP.11 Thus, the intraoperative use of PEEP cannot be seen as clinical standard.

Postoperative pulmonary complications, in particular after general anesthesia > 2 hours for abdominal surgery, add to the morbidity and mortality of surgical patients.12,13 We hypothesize that a lung–protective mechanical ventilation strategy with higher levels of PEEP and recruitment manoeuvres attenuates postoperative pulmonary complications in patients without lung injury (i.e., patients who do not fulfil the criteria for ALI at the moment of surgery).

PROVHILO aims at comparing the effects of such protective strategy and conventional mechanical ventilation in biomarkers of lung injury, postoperative pulmonary complications, extra pulmonary complications and length of hospital stay in patients undergoing general anesthesia for open abdominal surgery.

Methods

Objectives and designThe PROtective Ventilation using HIgh versus LOw positive end–expiratory pressure (“PROVHILO”) trial is a worldwide investigator-initiated multicenter randomized controlled two-arm trial.

The Institutional Review Board of the Academic Medical Center – University of Amsterdam, Amsterdam, The Netherlands, approved the trial. The PROVHILO trial is conducted in accordance

162

with the declaration of Helsinki and was registered on October 29 2010 at www.controlled-trials.com with trial identification number ISRCTN70332574.

Figure 1. CONSORT diagram of the PROVHILO trial

Study populationLocal investigators screen consecutive patients scheduled for non–laparoscopic abdominal surgery in participating centres worldwide. Demographic data on screened patients regardless of meeting enrolment criteria are recorded (registry: age, gender, type of surgery). A total of 900 patients are randomized to the 2 different mechanical ventilation strategies. In the participating centres at least 2 investigators are involved with the study. One researcher is involved with mechanical ventilation practice in the operation room, he/she will be blinded for the randomized intervention most closely to the time of tracheal intubation (depending on local situation) – the second investigator, blinded for randomization arm, will score the primary and secondary postoperative endpoints.

PROVHILO study design

Chap

ter

7

163

Patients with high or intermediate risk for postoperative pulmonary complications following non–laparoscopic abdominal surgery with general anesthesia are eligible for participation. To identify such patients the ARISCAT risk score (see Table 2) will be used.14 This predictive risk index is developed by the ARISCAT Group to assess the individual preoperative risk for post–operative pulmonary complications. An ARISCAT risk score ≥ 26 is associated with an intermediate to high risk for postoperative pulmonary complications.

Patients planned for laparoscopic surgery are excluded from participation, as are non–adult patients (age < 18 years), patients with a body mass index > 40 kg/m2, pregnant patients (excluded by laboratory analysis), and patients who consented for another interventional study or decline to participate. In addition, patients who were on mechanical ventilation > 30 minutes (e.g., because of general anesthesia for surgery) within last 30 days, are excluded. Other important exclusion criteria include: any previous lung surgery, history of previous severe chronic obstructive pulmonary disease (COPD) with (non–invasive) ventilation and/or oxygen therapy at home and/or repeated systemic corticosteroid therapy for acute exacerbations of COPD, ALI or acute respiratory distress syndrome expected to require prolonged post–operative mechanical ventilation, persistent hemodynamic instability or intractable shock (considered hemodynamic unsuitable for the study by the patient’s managing physician), severe cardiac disease (New York Heart Association class III or IV, or acute coronary syndrome, or persistent ventricular tachyarrhythmia’s), and recent immunosuppressive medication (receiving chemotherapy or radiation therapy within last 2 months).

All patients are asked for signed informed consent, as required by the institutional review board in accordance with the Declaration of Helsinki.

Randomization and interventionRandomization is performed using a dedicated, password protected, SSL-encrypted website. Randomization sequence is generated using random blocks and is stratified per centre. No blocking is applied to other trial factors.

Patients are randomly assigned to mechanical ventilation with levels of PEEP at 12 cmH2O with the use of recruitment manoeuvres (the lung–protective strategy) or mechanical ventilation with levels of PEEP at maximum 2 cmH2O without recruitment manoeuvres (the conventional strategy). The PEEP level in the protective strategy is chosen to be 12 cmH2O, to achieve maximal interventional effect without causing harm to participating patients and to make the intervention acceptable for the participating clinicians. The conventional strategy is chosen based on a national survey in France that showed > 90% of responding anaesthetists to use levels of PEEP of 0 – 4 cmH2O without recruitment maneuvers.11 Since not all available anesthesia ventilators can apply levels of PEEP < 2 cmH2O, the level of PEEP is set at a maximum of 2 cmH2O with the conventional strategy. However the lowest possible level of PEEP is always chosen.

Mechanical ventilationPatients are ventilated with a volume–controlled mechanical ventilation strategy. Although it is left to the discretion of the attending anaesthesiologist to use different fractions of inspired oxygen, it is advised to use at least 0.4, with the lowest oxygen fraction to maintain oxygen

164

saturation ≥ 92%. The inspiratory to expiratory time ratio (I:E) is set at 1:2, and the respiratory rate is adjusted to reach normocapnia (end–tidal carbon dioxide partial pressure between 35 and 45 mmHg). Tidal volumes of < 8 mL/kg predicted body weight (PBW) are advised to be used [15]. PBW is calculated according to a predefined formula: 50 + 0.91 x (centimetres of height – 152.4) for males and 45.5 + 0.91 x (centimetres of height – 152.4) for females.16,17 Tidal volumes throughout this protocol refer to the actual inspired tidal volume in the ventilator circuit.

Recruitment manoeuvreRecruitment manoeuvres, as part of the lung–protective strategy, are performed directly after intubation, after any disconnection from the mechanical ventilator, and directly before tracheal extubation. Recruitment manoeuvres should not be performed when patients are hemodynamic unstable, as judged by the attending physician.

Recruitment manoeuvres are not easily applied with available anesthesia ventilators since not all machines have an inspiratory hold function or other adequate facilities. To obtain standardization among centres, recruitment manoeuvres are performed as follows:

1. peak inspiratory pressure limit is set at 45 cmH2O

2. tidal volume is set at 8 ml/kg PBW and respiratory rate at 6-8 breaths/min (or lowest respiratory rate that anesthesia ventilator allows), while PEEP is set at 12 cmH2O

3. inspiratory to expiratory ratio (I:E) is set at 1:2

4. tidal volumes are increased in steps of 4 ml/kg PBW until a plateau pressure of 30–35 cmH2O

5. 3 breaths are administered with a plateau pressure of 30 – 35 cmH2O

6. peak inspiratory pressure limit, respiratory rate, I:E, and tidal volume are set back to settings preceding each recruitment manoeuvre, while maintaining PEEP at 12 cmH2O

Protocol drop–outAnesthesiologists are allowed to change the ventilation protocol at any time point upon the surgeon’s request, or if there is any concern about patient’s safety. The level of PEEP can be modified according to the anaesthesiologist in charge if the systolic arterial pressure drops < 90 mmHg for more than 3 minutes despite intravenous fluid infusion and/or start of vasopressors, if dosages of vasopressors are at the highest level tolerated, if new arrhythmias develop which are unresponsive to treatment suggested by the Advanced Cardiac Life Support Guidelines,18 if there is need of massive transfusion to maintain Ht > 21% (Hb > 7 mg/dl), or if there is a surgical complication determining life–threatening situations.

Rescue therapyIn both study groups, in case of desaturation (SpO2 < 90%), after excluding airway problems, severe hemodynamic impairment and ventilator malfunction, a rescue strategy is proposed, which improves oxygenation with respectively a decreasing level of PEEP with increasing FiO2 in

PROVHILO study design

Chap

ter

7

165

the lung–protective strategy group, and increasing levels of PEEP and FiO2 in the conventional group (see table 1).

Standard proceduresThe study protocol stresses that routine general anesthesia, postoperative pain management, physiotherapeutic procedures and fluid management must be used in the perioperative as well as the postoperative period according to each centres specific expertise and routine clinical use, to minimize interference with the trial intervention. However, it is suggested to perform postoperative pain management in order to achieve a visual analogue scale (VAS) pain score < 3, to use regional or neuroaxial analgesia if indicated, to use physiotherapy by early mobilization, deep breathing exercises with and without incentive spirometry, and stimulation of cough in the postoperative period, to avoid fluid overload (according to the discretion of the responsible physicians) and to use appropriate prophylactic antibiotics when indicated. Data on the applied procedures will be collected and analysed.

Table 1. Rescue therapies with the protective and the conventional strategy

Protective Conventional

Step FiO2 PEEP Step FiO2 PEEP

1 0.5 12 1 0.5 2

2 0.5 10 2 0.6 2

3 0.5 8 3 0.6 3

4 0.5 6 4 0.6 4

5 0.6 6 5 0.6 5

6 0.7 6 6 0.7 5

7 0.8 6 7 0.8 5

8 0.8 4 or lower 8 0.8 6

9 RM 6

PEEP: positive end–expiratory pressure; FiO2: fractional inspired oxygen; RM: recruitment manoeuvre

Follow upBaseline variables are collected pre–operative at the pre–anaesthetic visit or before induction of general anesthesia. The following variables are collected; gender, age, height, weight, functional status (independent, partially dependent or totally dependent), physical status (according to the American Society of Anesthesiologists (ASA), cardiac status (heart failure, according to the New York Heart Association (NYHA), acute coronary syndrome, or persistent ventricular tachyarrhythmia’s), COPD and use of inhalation therapy and/or steroids, respiratory infection in the last month, smoking status, alcohol status in the past 2 weeks, history of active cancer,

166

weight loss > 10% in the last 6 months, history of diabetes mellitus, use of oral anti–diabetics, use of antibiotics in the last 3 months, use of statins, type of scheduled surgery (emergency or non–emergency and surgical procedure), transfusion of blood products in the preceding 6 hours, vital parameters (tympanic temperature, respiratory rate, SpO2 (%), blood pressure, heart rate), airway secretion score (the patient is required to cough and the presence of secretion will be subjectively evaluated; if yes: purulent or not), VAS–scores for dyspnoea and pain, blood samples (glycaemia, uraemia, creatinine, AST, ALT, bilirubin, Hb, WBC count, platelet count, PT, PTT, and biomarkers [see below]) and a chest X–ray (assessed on mono- and bilateral infiltrate, pleural effusion, atelectasis, pneumothorax, cardiopulmonary oedema).

During the intraoperative period variables are recorded hourly after induction of anesthesia during the recruitment manoeuvre. These variables include duration and type of both anesthesia and surgical procedures, all administered drugs during anesthesia (e.g. anaesthetics, vasoactive drugs, anti-arrhythmic medication), ventilator settings, vital parameters, fluid– and transfusion requirements, need of rescue therapy for hypoxemia and intraoperative complications possibly related to recruitment manoeuvres (e.g. desaturation, hypotension during recruitment manoeuvre, need for vasoactive medication).

Patients are assessed at the first five postoperative days and at the last day before discharge from the hospital. On day 90 hospital free-days are recorded; if the patient is still admitted to the hospital on day 90, this day will be recorded as last day of follow-up. Clinical data and the presence of pulmonary and extra–pulmonary postoperative complications are scored; the day of development of any complication is indicated. A chest X–ray will be taken on the first post–operative day, blood samples for laboratory tests (glycaemia, uraemia, creatinine, AST, ALT, bilirubin, Hb, WBC count, platelet count, PT, PTT) will be taken on day 1, 3 and 5 and blood samples for biomarkers are collected directly after surgery and on day 5. As mentioned above, one local investigator, blinded for randomization group will score the primary and secondary postoperative endpoints.

Study endpointsPrimary endpoint – is a composed endpoint of all postoperative pulmonary complications with each complication weighing equally; it is presented as a total percentage of post–operative pulmonary complications. The postoperative complications are defined as: (a) mild respiratory failure (PaO2 < 60 mmHg or SpO2 < 90% in room air but responding to supplemental oxygen, (b) severe respiratory failure (need for non–invasive or invasive mechanical ventilation or a PaO2 < 60 mmHg or SpO2 < 90% despite supplemental oxygen), (c) development of ALI/ARDS (according to consensus guidelines),19 (d) suspected pulmonary infection (patient receives antibiotics and meets at least one of the following criteria: new or changed sputum, new or changed lung opacities on chest X–ray when clinically indicated, tympanic temperature > 38.30C, WBC count > 12,000/ml in the absence of other infectious focus), (e) pulmonary infiltrate (chest X–ray demonstrating unilateral or bilateral infiltrates), (f) pleural effusion (chest X–ray demonstrating blunting of the costophrenic angle, loss of the sharp silhouette of the ipsilateral hemidiaphragm in upright position, evidence of displacement of adjacent anatomical structures or (in supine position) a hazy opacity in one hemi–thorax with preserved vascular shadows), (g) atelectasis (suggested by lung opacification with shift of the mediastinum, hilum, or hemidiaphragm towards the affected

PROVHILO study design

Chap

ter

7

167

Table 2. ARISCAT risk score Independent predictors of risk for post-operative pulmonary complications identified in the logistic regression model

Multivariate ModelOR (95% CI)n = 1,624

β Coefficient Risk Score†

Age (years)

≤ 50 1

51 – 80 1.4 (0.6 – 3.3) 0.331 3

> 80 5.1 (1.9 – 13.3) 1.619 16

Pre-operative (SpO2, %)

≥ 96 1

91 – 95 2.2 (1.2 – 4.2) 0.802 8

≤ 90 10.7 (4.1 – 28.1) 2.375 24

Respiratory infection 5.5 (2.6 – 11.5) 1.698 17

in the last month

Pre-operative anaemia (≤ 10 g/dl) 3.0 (1.4 – 6.5) 1.105 11

Surgical incision

Peripheral 1

Upper abdominal 4.4 (2.3 – 8.5) 1.480 15

Intra-thoracic 11.4 (4.9 – 26.0) 2.431 24

Duration of surgery (hours)

≤ 2 1

2 – 3 4.9 (2.4 – 10.1) 1.593 16

> 3 9.7 (4.7 – 19.9) 2.268 23

Emergency procedure 2.2 (1.0 – 4.5) 0.768 8

High or intermediate risk for postoperative pulmonary complications ≥ 26

CI: confidence interval; OR: odds ratio; SpO2: oxyhemoglobin saturation by pulse oximetry breathing air in supine position; g/dL: gram per decilitre †The simplified risk score was the sum of each logistic regression coefficient multiplied by 10, after rounding off its value

area, and compensatory overinflation in the adjacent non-atelectatic lung), (h) pneumothorax (air in the pleural space with no vascular bed surrounding the visceral pleura), (i) bronchospasm (newly detected expiratory wheezing treated with bronchodilators), (j) aspiration pneumonitis (respiratory failure after the inhalation of regurgitated gastric contents), (k) cardiopulmonary

168

oedema (clinical signs of congestion, including dyspnoea, oedema, rales and jugular venous distention, with the chest X–ray demonstrating increase in vascular markings and diffuse alveolar interstitial infiltrates).

Secondary clinical endpoints – include (a) intraoperative ventilation strategy related complications (e.g. desaturation, hypotension during recruitment manoeuvre, need for vasoactive medication), (b) unexpected need for ICU admission or ICU readmission, (c) hospital–free days at follow–up day 90, (d) post–operative wound healing and (e) post–operative extra–pulmonary complications. Extra–pulmonary complications include SIRS, sepsis, severe sepsis, septic shock (all according to consensus criteria),20 extra–pulmonary infection (wound infection or any other infection), coma (Glasgow Coma Score < 8 in the absence of therapeutic coma or sedation), acute myocardial infarction (according to universal definition of myocardial infarction),21 acute renal failure (according to the RIFLE classification system),22 disseminated intravascular coagulation (according to ISTH diagnostic scoring system for DIC),23 gastro–intestinal failure (defined as; gastro-intestinal bleeding or gastro-intestinal failure according to GIF–score)24 and hepatic failure (defined as; serum bilirubin level > 2 mg/dL with elevation of the transaminase and lactic dehydrogenase levels above twice normal values).

Other study parameters – Blood samples will be collected and analysed for systemic markers of lung injury (including but not limited to soluble Receptor for Advanced Glycation Endproducts (sRAGE), Clara Cell protein–16 (CC–16), surfactant proteins A and D and levels of proinflammatory and procoagulant/antifibrinolytic mediators (including but not limited to interleukin (IL)–6, IL–8, tumor necrosis factor (TNF)–α, and thrombin–antithrombin (TAT), protein C, and plasminogen activator inhibitor (PAI)–1). The abovementioned biomarkers of lung injury, acute inflammation and coagulopathy have been shown to correlate with poor clinical outcome in patients with ALI/ARDS.25 Notably, with short term mechanical ventilation rises in systemic levels of lung injury biomarkers,26 acute inflammation9 and procoagulant/ antifibrinolytic mediators8 have been described. Lung–protective mechanical ventilation strategies attenuated the rise in levels of some of the abovementioned mediators in patients with ALI/ARDS,27 as well as patients who underwent short term mechanical ventilation because of surgery.8,9 Most of these trials compared the effect of different tidal volumes.

The injury induced by mechanical ventilation originates in the lung, but may also affect distal organs by release of mediators from the lung into the systemic circulation.28,29 Therefore systemic biomarkers of distant organ injury, in particular the kidney, are determined (including, but not limited to neutrophil gelatinase-associated lipocalin (NGAL) and cystatin C).

Statistical considerationsSample size calculation – the required sample size is calculated from an estimated effect size derived from data collected in the ARISCAT study14 and previous studies on the incidence of postoperative pulmonary complications.12,13,30 A two group χ2 test with a 0.05 two–sided significance level will have 80% power to detect the difference (in post–operative pulmonary complications) between conventional mechanical ventilation (24%) and open lung mechanical ventilation (16.5%) (Odds ratio of 0.626) when the sample size in each group is 450.

PROVHILO study design

Chap

ter

7

169

Interim analysis – one main concern is not to withhold positive effects of the open lung mechanical ventilation strategy to the control group. Therefore, interim analyses are performed after 300 and 600 patients. The first interim analysis is performed when 300 patients have successfully been included and followed-up. If the intervention has a strong trend for improving postoperative pulmonary complications (as defined above) with a p–value < 0.0005 is found at 300 patients or < 0.014 at 600 patients, termination of the study is considered. The third and final analysis is performed at 900 patients with a p–value of 0.045 for significance. When postoperative pulmonary complications occur significantly more frequent in the intervention group, terminating the study due to harm will be considered when p ≤ 0.022 for each interim analysis.

Statistical analysisNormally distributed variables will be expressed by their mean and standard deviation; not normally distributed variables will be expressed by their medians and interquartile ranges; categorical variables will be expressed as n (%). In test groups of continuous normally distributed variables, Student’s t–test will be used. Likewise if continuous data are not normally distributed the Mann–Whitney U test will be used. Categorical variables will be compared with the Chi–square test or Fisher’s exact tests or when appropriate as relative risks. Where appropriate statistical uncertainty will be expressed by 95% confidence levels.

Primary outcome is the total occurrence of pulmonary complications within the first 5 post–operative days, presented as a percentage. The percentage will be analysed as continuous data. If the data is normally distributed, Student’s t–test will be used or when not normally distributed the Mann–Whitney U test will be used.

As this is a randomized controlled trial, we expect that randomization in this large study population will sufficiently balance the baseline characteristics. Baseline balance is tested and imbalance compensated in all pre-operative variables and on ARISCAT scores (as mentioned above).14 However if imbalance occurs, the confounding factor will be corrected using a multiple logistic regression model. For this we will treat the proportion as a binary response (complications occur during day one to day five postoperative).

Time to event variables (primary and secondary outcomes) are analysed using a proportional hazard model adjusted for possible imbalances of patients’ baseline characteristics. Time course variables (e.g. repeated measures of vital parameters, blood values, VAS–scores, actual mobility) are analysed by a linear mixed model. The linear mixed models procedure expands the GLM so that the data are permitted to exhibit correlated and non–constant variability. The model includes two factors: 1) study group (fixed factor, intervention or control group), each level of the study group factor can have a different linear effect on the value of the dependent variable; 2) time as a covariate, time is considered to be a random sample from a larger population of values, the effect is not limited to the chosen times.

Study OrganizationThe Executive Committee is constituted of the study principal investigator and the principal investigators of the investigating centres that approved the final trial design and protocol issued

170

to the clinical sites and to the Data and Safety Monitoring Board (DSMB).

The independent DSMB watches over the ethics of conducting the study in accordance with the Declaration of Helsinki, monitors patient safety and reviews safety issues as the study progresses. All serious adverse events, and all unexpected and related or possibly–related adverse events will be reported blinded to the appointed international SAE–manager, who assesses the events and reports this information to the DSMB within 24 hours of that event in the case of a serious adverse event or within one week in the case of an adverse event.

The Steering Committee is composed of the principal investigators of the principle participating centres that contribute to the design and revisions of the study protocol.

The National Coordinators are responsible for administrative management and communication with the local principle investigator and provide assistance to the participating clinical sites in trial management, record keeping and data management.

Discussion

It has become clear that mechanical ventilation can attenuate lung damage and may even be the primary factor in lung injury.2,3 ALI/ARDS is characterized by heterogeneous distribution of pulmonary aeration. During ventilation the aerated part of the lung receives the largest part of the tidal volume, potentially causing overdistention with excessive alveolar wall tension and stress. The non–aerated atelectatic lung regions are prone to repeated collapse and re–expansion of alveoli, causing shear stress and diffuse mechanical damage of the alveoli.2,28 This could trigger local and systemic inflammation, which has been suggested to cause ventilator–associated lung injury.1,8,9

Protective mechanical ventilation using lower tidal volumes could reduce ventilator–associated lung injury. Indeed, the use of lower tidal volumes has been found beneficial in patients who needed long–term mechanical ventilation for ALI/ARDS.1,15 Two retrospective studies31,32 and one randomized controlled trial33 suggest lower tidal volumes to be beneficial in patients without acute lung injury in long-term ventilation as well. Other trials suggest that ventilation with lower tidal volumes is also beneficial in short term ventilation for patients without preexisting lung injury.8,9,34 In these trials different levels of PEEP were used, making comparison and interpretation of the additional effect of PEEP difficult.

During general anesthesia reductions in end–expiratory lung volume and increases in airway closure is commonly seen.35 Both contribute to atelectasis formation. The most important morbid postoperative pulmonary complication is atelectasis formation, which increases the risk for pneumonia and hypoxic acute respiratory failure.36 Postoperative pulmonary complications, in particular postoperative respiratory failure, add to the morbidity and mortality of surgical patients.12,13 PEEP prevents alveolar collapse and atelectasis formation. Recruitment manoeuvres can be used to achieve initial alveolar recruitment.3,37 Data suggests that recruitment manoeuvres

PROVHILO study design

Chap

ter

7

171

adequately support the beneficial effects of PEEP in short-term ventilation.38,39 However, PEEP levels should not be too high, to avoid overdistention of the lung.1,40

Various studies showed mechanical ventilation according to an open lung concept to improve ventilatory efficacy of the lungs in patients with healthy lungs undergoing general anesthesia.3,37 Studies have shown the open lung concept to attenuate inflammatory responses and to prevent loss of functional residual capacity in cardiac surgery patients.34,41 Of note, there is some controversy about the clinical importance of the cyclic collapse of alveoli. Indeed, the potential of ventilation strategies with lower tidal volumes and PEEP for protecting the lungs during the intraoperative period in patients without previous lung injury has been questioned.6,7

The PROVHILO trial is the first randomized controlled study powered to investigate whether protective mechanical ventilation using higher levels of PEEP complemented by recruitment manoeuvres attenuates postoperative pulmonary complications. The two ventilation strategies used in the PROVHILO trial are composed to match as many clinically applied anesthesia ventilators as possible. With these standardized ventilation strategies, we aim to minimize variation between ventilation strategies used in the participating centres.

The primary endpoint of this trial is a composed endpoint (postoperative pulmonary complications). This could be seen as a shortcoming, since the effect of the intervention on one postoperative pulmonary complication could be diluted if other postoperative pulmonary complications are not affected, or affected to a lesser content. However, since we collect and report on all postoperative pulmonary complications, it may still be possible to determine the effects on separate complications.

The main concern in the statistical interim analysis is not to withhold positive effects of the treatment to the control group. However, to achieve maximal protection for patients and to have a lower chance of achieving positive effects of the intervention on postoperative pulmonary complications if they were not really present, different stopping rules are defined for a strong beneficial effect on postoperative pulmonary complications of the intervention versus a worse effect on postoperative pulmonary complications.

The spectrum of ventilator–associated lung injury does not only include pulmonary inflammation, but also an increase in systemic inflammatory mediators.2,42-44 The lung has been suggested as an important causative part of the inflammation–induced systemic disease state that can evolve to multi organ failure, rather than merely a pulmonary disease process. Alveolar collapse during mechanical ventilation can lead to activation of inflammatory response both locally and systemically, which can play a role in modulating the individual patient’s outcome.3,28,45 To determine this possible effect on patients in this trial, secondary endpoints on extra–pulmonary complications are collected and reported, as well as blood samples for the determination of specific markers of distal organ injury.

Several confounding factors can be suggested. Postoperative pain is a commonly acknowledged contributor to postoperative atelectasis.46,47 Respiratory chest physiotherapy has been shown to decrease postoperative respiratory complications in cardiac surgery, when performed before

172

surgery.48 It is still uncertain if postoperative physiotherapeutic procedures are beneficial, although there is some evidence in favour of physiotherapy.47 Excessive intraoperative fluid administration is another possible contributing factor to the development of respiratory failure.49 These factors are not protocolized by the PROVHILO trial. The protocol stresses that general anesthesia, post–operative pain management, physiotherapeutic procedures, fluid management and all other perioperative procedures are to be performed according to the centres’ specific expertise and routine clinical use. We aim to minimize interference with the effect of PEEP and recruitment manoeuvres on postoperative pulmonary complications. Suggestions on the abovementioned perioperative procedures are made in the protocol, to keep the variability as small as possible. No suggestions are made on type of anesthesia to use, to make the trial as accessible as possible for anaesthesiologists. It is known, however, that several anaesthetic drugs affect lung capacity during surgery.50,51 Since we collect and report on all commonly known risk factors for postoperative pulmonary complications and intra-operative administered drugs, it may still be possible to determine the effect on the primary and secondary outcomes.

In conclusion, the PROVHILO trial is a worldwide investigator–initiated randomized controlled trial powered to test the hypothesis that an open lung mechanical ventilation strategy using higher levels of PEEP and recruitment manoeuvres during short term intraoperative mechanical ventilation prevents against post–operative pulmonary complications. The PROVHILO trial also determines the effect of an open lung approach on postoperative extra–pulmonary complications. Finally, in the PROVHILO trial the effect of lung–protective mechanical ventilation is monitored by highly specific biomarkers of lung injury.

Funding Source

This study is an investigator–initiated trial, funded by the Academic Medical Center at the University of Amsterdam, Amsterdam, The Netherlands, and the European Society of Anesthesiology (ESA).

PROVHILO study design

Chap

ter

7

173

References1. Putensen C, Theuerkauf N, Zinserling J, Wrigge H, Pelosi P. Metaanalysis: ventilation strategies and outcomes of the

acute respiratory distress syndrome and acute lung injury. Ann Intern Med 2009, 151:566-5762. Tremblay LN, Slutsky AS. Ventilator-induced lung injury: from the bench to the bedside. Intensive Care Med 2006, 32:24-333. Papadakos PJ, Lachmann B. The open lung concept of mechanical ventilation: the role of recruitment and stabilization.

Crit Care Clin 2007, 23:241-250, ix-x4. Briel M, Meade M, Mercat A, Brower RG, Talmor D, Walter SD, Slutsky AS, Pullenayegum E, Zhou Q, Cook D, et al.

Higher vs lower positive end-expiratory pressure in patients with acute lung injury and acute respiratory distress syndrome: systematic review and metaanalysis. JAMA, 303:865-873

5. Schultz MJ, Haitsma JJ, Slutsky AS, Gajic O. What tidal volumes should be used in patients without acute lung injury? Anesthesiology 2007, 106:1226-1231

6. Wrigge H, Uhlig U, Baumgarten G, Menzenbach J, Zinserling J, Ernst M, Dromann D, Welz A, Uhlig S, Putensen C. Mechanical ventilation strategies and inflammatory responses to cardiac surgery: a prospective randomized clinical trial. Intensive Care Med 2005, 31:1379-1387

7. Wrigge H, Uhlig U, Zinserling J, Behrends-Callsen E, Ottersbach G, Fischer M, Uhlig S, Putensen C. The effects of different ventilatory settings on pulmonary and systemic inflammatory responses during major surgery. Anesth Analg 2004, 98:775-781

8. Choi G, Wolthuis EK, Bresser P, Levi M, van der Poll T, Dzoljic M, Vroom MB, Schultz MJ. Mechanical ventilation with lower tidal volumes and positive end-expiratory pressure prevents alveolar coagulation in patients without lung injury. Anesthesiology 2006, 105:689-695

9. Wolthuis EK, Choi G, Dessing MC, Bresser P, Lutter R, Dzoljic M, van der Poll T, Vroom MB, Hollmann M, Schultz MJ: Mechanical ventilation with lower tidal volumes and positive end-expiratory pressure prevents pulmonary inflammation in patients without preexisting lung injury. Anesthesiology 2008, 108:46-54

10. Rothen HU, Sporre B, Engberg G, Wegenius G, Reber A, Hedenstierna G. Prevention of atelectasis during general anaesthesia. Lancet 1995, 345:1387-1391

11. Jaber S, Coisel Y, Marret E, Malinovsky JM, Bouaziz H. Ventilatory Management during General Anesthesia: A Multicenter Observational Study. Anesthesiology 2006:A1516

12. Arozullah AM, Daley J, Henderson WG, Khuri SF. Multifactorial risk index for predicting postoperative respiratory failure in men after major noncardiac surgery. The National Veterans Administration Surgical Quality Improvement Program. Ann Surg 2000, 232:242-253

13. Smetana GW, Lawrence VA, Cornell JE. Preoperative pulmonary risk stratification for noncardiothoracic surgery: systematic review for the American College of Physicians. Ann Intern Med 2006, 144:581-595

14. Canet J, Gallart L, Gomar C, Paluzie G, Valles J, Castillo J, Sabate S, Mazo V, Briones Z, Sanchis J. Prediction of Postoperative Pulmonary Complications in a Population based Surgical Cohort. Anesthesiology, 113:1338-1350

15. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. The Acute Respiratory Distress Syndrome Network. NEJM 2000, 342:1301-1308

16. Crapo RO, Morris AH, Gardner RM. Reference spirometric values using techniques and equipment that meet ATS recommendations. Am Rev Respir Dis 1981, 123:659-664

17. Crapo RO, Morris AH, Clayton PD, Nixon CR. Lung volumes in healthy nonsmoking adults. Bull Eur Physiopathol Respir 1982, 18:419-425

18. 2005 International Consensus on Cardiopulmonary Resuscitation and Emergency Cardiovascular Care Science with Treatment Recommendations. Part 4: Advanced life support. Resuscitation 2005, 67:213-247

19. Bernard GR, Artigas A, Brigham KL, Carlet J, Falke K, Hudson L, Lamy M, LeGall JR, Morris A, Spragg R. Report of the American-European consensus conference on ARDS: definitions, mechanisms, relevant outcomes and clinical trial coordination. The Consensus Committee. Intensive Care Med 1994, 20:225-232

20. Bone RC. Toward an epidemiology and natural history of SIRS (systemic inflammatory response syndrome). JAMA 1992, 268:3452-3455

21. Thygesen K, Alpert JS, White HD, Jaffe AS, Apple FS, Galvani M, Katus HA, Newby LK, Ravkilde J, Chaitman B, et al. Universal definition of myocardial infarction. Circulation 2007, 116:2634-2653

22. Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky P. Acute renal failure – definition, outcome measures, animal models, fluid therapy and information technology needs: the Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care 2004, 8:R204-212

23. Levi M, Toh CH, Thachil J, Watson HG. Guidelines for the diagnosis and management of disseminated intravascular coagulation. British Committee for Standards in Haematology. Br J Haematol 2009, 145:24-33

24. Reintam A, Parm P, Kitus R, Starkopf J, Kern H. Gastrointestinal failure score in critically ill patients: a prospective observational study. Crit Care 2008, 12:R90

25. Levitt JE, Gould MK, Ware LB, Matthay MA. The pathogenetic and prognostic value of biologic markers in acute lung

174

injury. J Intensive Care Med 2009, 24:151-16726. Determann RM, Wolthuis EK, Choi G, Bresser P, Bernard A, Lutter R, Schultz MJ. Lung epithelial injury markers are

not influenced by use of lower tidal volumes during elective surgery in patients without preexisting lung injury. Am J Physiol Lung Cell Mol Physiol 2008, 294:L344-350

27. Parsons PE, Eisner MD, Thompson BT, Matthay MA, Ancukiewicz M, Bernard GR, Wheeler AP. Lower tidal volume ventilation and plasma cytokine markers of inflammation in patients with acute lung injury. Crit Care Med 2005, 33:1-6

28. Del Sorbo L, Slutsky AS. Ventilatory support for acute respiratory failure: new and ongoing pathophysiological, diagnostic and therapeutic developments. Curr Opin Crit Care, 16:1-7

29. Plotz FB, Slutsky AS, van Vught AJ, Heijnen CJ. Ventilator-induced lung injury and multiple system organ failure: a critical review of facts and hypotheses. Intensive Care Med 2004, 30:1865-1872

30. Arozullah AM, Khuri SF, Henderson WG, Daley J. Development and validation of a multifactorial risk index for predicting postoperative pneumonia after major noncardiac surgery. Ann Intern Med 2001, 135:847-857

31. Gajic O, Dara SI, Mendez JL, Adesanya AO, Festic E, Caples SM, Rana R, St Sauver JL, Lymp JF, Afessa B, Hubmayr RD. Ventilator-associated lung injury in patients without acute lung injury at the onset of mechanical ventilation. Crit Care Med 2004, 32:1817-1824

32. Gajic O, Frutos-Vivar F, Esteban A, Hubmayr RD, Anzueto A. Ventilator settings as a risk factor for acute respiratory distress syndrome in mechanically ventilated patients. Intensive Care Med 2005, 31:922-926

33. Determann RM, Royakkers A, Wolthuis EK, Vlaar AP, Choi G, Paulus F, Hofstra JJ, de Graaff MJ, Korevaar JC, Schultz MJ. Ventilation with lower tidal volumes as compared with conventional tidal volumes for patients without acute lung injury: a preventive randomized controlled trial. Crit Care 2010, 14:R1

34. Reis Miranda D, Gommers D, Struijs A, Dekker R, Mekel J, Feelders R, Lachmann B, Bogers AJ. Ventilation according to the open lung concept attenuates pulmonary inflammatory response in cardiac surgery. Eur J Cardiothorac Surg 2005, 28:889-895

35. Pelosi P, Rocco PR. Airway closure: the silent killer of peripheral airways. Crit Care 2007, 11:11436. Pelosi P, Jaber S. Noninvasive respiratory support in the perioperative period. Curr Opin Anaesthesiol, 23:233-23837. Lapinsky SE, Mehta S. Bench-to-bedside review: Recruitment and recruiting maneuvers. Crit Care 2005, 9:60-6538. Girgis K, Hamed H, Khater Y, Kacmarek RM. A decremental PEEP trial identifies the PEEP level that maintains oxygenation

after lung recruitment. Respir Care 2006, 51:1132-113939. Talab HF, Zabani IA, Abdelrahman HS, Bukhari WL, Mamoun I, Ashour MA, Sadeq BB, El Sayed SI. Intraoperative

ventilatory strategies for prevention of pulmonary atelectasis in obese patients undergoing laparoscopic bariatric surgery. Anesth Analg 2009, 109:1511-1516

40. Maisch S, Reissmann H, Fuellekrug B, Weismann D, Rutkowski T, Tusman G, Bohm SH. Compliance and dead space fraction indicate an optimal level of positive end-expiratory pressure after recruitment in anesthetized patients. Anesth Analg 2008, 106:175-181

41. Reis Miranda D, Struijs A, Koetsier P, van Thiel R, Schepp R, Hop W, Klein J, Lachmann B, Bogers AJ, Gommers D. Open lung ventilation improves functional residual capacity after extubation in cardiac surgery. Crit Care Med 2005, 33:2253-2258

42. Slutsky AS, Tremblay LN. Multiple system organ failure. Is mechanical ventilation a contributing factor? Am J Respir Crit Care Med 1998, 157:1721-1725

43. Imai Y, Parodo J, Kajikawa O, de Perrot M, Fischer S, Edwards V, Cutz E, Liu M, Keshavjee S, Martin TR, et al. Injurious mechanical ventilation and end-organ epithelial cell apoptosis and organ dysfunction in an experimental model of acute respiratory distress syndrome. JAMA 2003, 289:2104-2112

44. Dreyfuss D, Saumon G. Ventilator-induced lung injury: lessons from experimental studies. Am J Respir Crit Care Med 1998, 157:294-323

45. Papadakos PJ. Cytokines, genes, and ARDS. Chest 2002, 121:1391-139246. Block BM, Liu SS, Rowlingson AJ, Cowan AR, Cowan JA, Jr., Wu CL. Efficacy of postoperative epidural analgesia: a

metaanalysis. JAMA 2003, 290:2455-246347. Ferreyra G, Long Y, Ranieri VM. Respiratory complications after major surgery. Curr Opin Crit Care 2009, 15:342-34848. Filsoufi F, Rahmanian PB, Castillo JG, Chikwe J, Adams DH. Predictors and early and late outcomes of respiratory failure

in contemporary cardiac surgery. Chest 2008, 133:713-72149. Fernandez-Perez ER, Keegan MT, Brown DR, Hubmayr RD, Gajic O. Intraoperative tidal volume as a risk factor for

respiratory failure after pneumonectomy. Anesthesiology 2006, 105:14-1850. Chawla G, Drummond GB. Fentanyl decreases end-expiratory lung volume in patients anaesthetized with sevoflurane.

Br J Anaesth 2008, 100:411-41451. Hedenstierna G, Edmark L: The effects of anesthesia and muscle paralysis on the respiratory system. Intensive Care

Med 2005, 31:1327-1335

PROVHILO study design

Chap

ter

7

175

Chapter 8

High versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial

Hemmes SNT, Gama de Abreu M, Pelosi P, Schultz MJ for the PROVE Network Investigators for the Clinical Trial Network of the European Society of AnaesthesiologyLancet 2014; 9; 384(9942):495-503

178

Abstract

Background. The role of positive end–expiratory pressure (PEEP) in mechanical ventilation during general anaesthesia for surgery remains uncertain. Higher PEEP levels may protect against postoperative pulmonary complications (PPCs), but could also cause intra–operative circulatory depression and lung injury from overdistension. We tested the hypothesis that higher PEEP plus recruitment manoeuvres protects against PPCs in patients at risk for PPCs receiving mechanical ventilation with lower tidal volumes during general anaesthesia for open abdominal surgery.

Methods. In this international, randomized controlled trial in 30 centres we allocated 900 patients at risk for PPCs planned for open abdominal surgery under general anaesthesia and ventilation at tidal volumes of 8 ml/kg to higher PEEP (12 cmH2O) with recruitment manoeuvres or lower PEEP (≤ 2 cmH2O) without recruitment manoeuvres, using a centralized computer-generated randomization system. Patients and outcome assessors were blinded for the intervention. Primary endpoint was a composite of PPCs by postoperative day five. The study is registered at Controlled-Trials.gov, number ISRCTN70332574.

Findings. From February 2011 through January 2013, 447 patients were randomized to the higher PEEP and 453 to the lower PEEP group. Six patients were excluded from analysis: 4 withdrew consent, 2 violated inclusion criteria. Median PEEP levels were 12 [12–12] and 2 [0–2] cmH2O in the higher and lower PEEP group, respectively. PPCs occurred in 174 of 445 patients (40%) in the higher PEEP group and in 172 of 449 patients (39%) in the lower PEEP group (relative risk, 1.01; 95% CI 0.86–1.20; P = 0.86). In the higher PEEP group, patients developed intraoperative hypotension and needed more vasoactive drugs.

Interpretation. A strategy with higher PEEP plus recruitment manoeuvres during open abdominal surgery does not protect against PPCs.

Funding. Funded by the Academic Medical Center, Amsterdam, The Netherlands, and the European Society of Anaesthesiology.

PROVHILO trial

Chap

ter

8

179

Introduction

Postoperative pulmonary complications (PPCs) are at least as frequent as cardiac complications during non–cardiac surgery.1 PPCs increase mortality after open abdominal surgery.2,3 It is gradually more recognized that mechanical ventilation may influence the incidence of PPCs4 and even distal organ dysfunction.5 Different mechanisms have been proposed to explain the injurious effects of ventilation. Both hyperinflation and repetitive tidal recruitment of lung units can induce the release of pro–inflammatory mediators, leading to lung and distal organ injury.6

Prevention of hyperinflation by use of lower tidal volumes (VTs) reduces mortality in patients with the acute respiratory distress syndrome (ARDS).7 Prevention of repetitive tidal recruitment by use of higher positive end–expiratory pressure (PEEP) is also associated with reduced mortality in these patients, but only when ARDS is severe.8 Several studies suggest that use of lower VTs in patients without lung injury under general anaesthesia may also reduce the incidence of PPCs.4 This hypothesis was recently confirmed in one single–centre9 and one national multicentre trial.10 However, in both trials, the use of lower VTs was combined with higher PEEP, thus it is unclear whether beneficial effects came from prevention of hyperinflation or prevention of repetitive tidal recruitment. While it is argued that use of too low PEEP could lead to atelectasis with ventilation strategies that use lower VTs,6,11 higher PEEP could also provoke complications including intraoperative circulatory depression12 and even promote hyperinflation.13

We conducted the PROtective Ventilation using HIgh versus LOw PEEP (PROVHILO) trial to test the hypothesis that a ventilation strategy with higher levels of PEEP plus recruitment manoeuvres during general anaesthesia for open abdominal surgery protects against PPCs in patients at risk for PPCs.

Methods

The PROVHILO trial was an investigator–initiated, international, multicentre, double–blind, parallel randomized controlled two–arm trial. The study protocol and the statistical analysis plan were approved by the Institutional Review Boards of the Academic Medical Center (AMC), Amsterdam, The Netherlands, as well as of all participating centres and published both in TRIALS14 and Controlled Trials.gov, number ISRCTN70332574. The steering committee was responsible for accuracy and completeness of fidelity of the study to the protocol, the collected data and data analyses. The writing committee drafted the manuscript without editorial assistance and all authors provided revisions and comments.

An independent data safety and monitoring board oversaw conduct of the trial, safety of the participants, and interpreted blind interim analysis results. A random sample of 6 centres was visited on site by an independent monitor to assess protocol adherence. There was no industry support or involvement for the PROVHILO trial.

180

PatientsPatients were screened and randomized from February 2011 to January 2013 at 30 hospitals in ten countries. Participating hospitals are listed in the Supplementary Appendix. Written informed consent was obtained from all participating patients before randomization. We considered adults scheduled for open abdominal surgery under general anaesthesia. Enrolment was restricted to patients who had an intermediate or high risk of experiencing PPCs.3 Patients who were planned for laparoscopic surgery, were pregnant (excluded by laboratory analysis), had a body mass index > 40 kg/m2, had severe cardio– or pulmonary comorbidities or another condition that may have compromised safe trial procedure, or who had consented for another interventional study or declined to participate, were excluded from study participation. The full inclusion and exclusion criteria are presented in tables S1 and S2 in the Supplementary Appendix.

Randomization and maskingPatients were assigned to their study group in random blocks of four and stratified per centre. Local investigators randomized patients after inclusion of the patients, using a secured, centralized, computer–generated and web–based, randomization system. In each centre at least two investigators were involved: one who was aware of the allocated intervention and collected intraoperative data; the other remained blinded to the intraoperative interventions and evaluated the outcomes, scoring postoperative pulmonary and extrapulmonary complications. The allocation was also concealed from patients, research staff, the independent statistician, and the data safety and monitoring board. Data were collected on paper case report forms and transcribed by local investigators onto secure web–based electronic case report forms (OpenClinica, Boston, MA, USA).

InterventionsPatients were randomized to receive intraoperative ventilation using either a higher PEEP strategy, with PEEP of 12 cmH2O plus recruitment manoeuvres, or a lower PEEP strategy, with PEEP of ≤ 2 cmH2O without recruitment manoeuvres. In the higher PEEP group, recruitment manoeuvres with an incremental tidal volume strategy were performed directly after induction of anaesthesia, after any disconnection from the ventilator, and just before tracheal extubation (see table S3 for details). A rescue strategy was designated for patients in whom pulse oximetry (SpO2) decreased to < 90% without evidence of airway problems, severe hemodynamic impairment, or ventilator malfunction. The strategy included a stepwise increase of inspired oxygen (FIO2), progressive increase in PEEP and recruitment manoeuvres (table S3). It was sequentially implemented to return SpO2 to ≥ 92%.

Patients were ventilated using a volume–assist mode during surgery, and optionally switched to pressure support mode near the end of surgery. VTs were set at 8 ml/kg predicted body weight. FIO2 was set at 0.40 or higher to a target SpO2 ≥ 92%. The respiratory rate was adjusted to maintain end–tidal CO2 (FE’CO2) between 35 and 45 mmHg. The inspiration–to–expiration ratio was 1:2. Anaesthesiologists were allowed to change the ventilator settings upon the surgeon’s request, or if there was any concern about patient’s safety. Safety concerns potentially included low systemic blood pressure unresponsive to intravenous fluids and/or vasoactive drugs, new arrhythmias not responding to treatment, or a need for massive transfusion. Other aspects of general anaesthesia, fluid administration, and pain management were per routine.

PROVHILO trial

Chap

ter

8

181

OutcomesA collapsed composite of PPCs was chosen as the primary endpoint. PPCs occurring within the first five postoperative days included hypoxemia, severe hypoxemia, bronchospasm, suspected pulmonary infection, pulmonary infiltrate, aspiration pneumonitis, development of ARDS, atelectasis, pleural effusion, pulmonary oedema caused by cardiac failure and pneumothorax. Definitions are presented in table S4.

Intraoperative complications served as secondary and safety endpoints, and included SpO2 < 90% requiring rescue, hypotension (systolic arterial blood pressure < 90 mmHg for more than 3 minutes), any need for vasoactive medication, any new arrhythmias requiring intervention, massive transfusion (> 5 units of packed red blood cells during one hour), and any surgical complication. Postoperative extrapulmonary complications included development of systemic inflammatory response syndrome (SIRS), sepsis, severe sepsis or septic shock, extrapulmonary infection, coma, acute myocardial infarction, acute renal failure, disseminated intravascular coagulation, hepatic failure, gastro–intestinal bleeding, gastro–intestinal failure, and impaired wound healing (definitions are presented in table S5).

Patients were assessed daily, scoring clinical data, the presence of the predefined outcomes, and need for intensive care unit admission or readmission until the fifth postoperative day, and shortly before hospital discharge. Ninety days after surgery, we determined the number of hospital–free days (including admissions to other hospitals) and vital status.

Statistical analysisWe calculated that a sample size of 900 patients would provide a power of 80% to detect a difference between the incidences of PPCs with lower PEEP (24%) and higher PEEP (16.5%).1,3,9,15,16 An independent data safety and monitoring board conducted an interim analysis after enrolment of the first 300 and 600 patients, according to the a priori statistical analysis plan. The Board did not recommend trial discontinuation after either interim analysis and 900 patients were therefore included. All patients were analysed under intention–to–treat rules.

Postoperative variables were compared using Student’s t–test or Mann–Whitney U test for continuous variables depending on the characteristics of the variables and chi-square test for categorical variables. The composite primary outcome, total occurrence of PPCs, and the secondary outcome of total occurrence of extrapulmonary complications, both in the first five postoperative days, were compared using an unadjusted chi–square test weighing each individual complication equally. The primary endpoint was not adjusted for baseline imbalance. Due to the two interim analyses a two–sided alpha level of 0.045 was considered statistically significant for the primary endpoint. Statistical significance for other variables was accepted at a P–value < 0.05. Where appropriate, statistical uncertainty was expressed by 95% confidence levels. Kaplan–Meier estimates of survival curves were calculated; log–rank tests were used to compare survival distributions between the lower and higher PEEP group. Data used for the Kaplan–Meier estimates was censored when patients did not experience a PPC during the study period, or when patients were lost to follow up before the end of day five. A post–hoc analysis was performed on the primary endpoint, discarding hypoxemia from the composite endpoint of PPCs. Further exploratory post–hoc analyses included a per–protocol analysis in which patients

182

of the higher PEEP group who did not receive higher PEEP throughout the procedure and all RMs as indicated by the study protocol were analysed as patients in the lower PEEP group, and the association between the incidence of postoperative complications was analysed; intra–operative use of medication (anaesthetics, neuromuscular blocking agents, and opioids); the net effect of the treatment group (higher PEEP) on the primary endpoint (PPCs), controlling for centre; and a multiple logistic regression analysis to identify baseline and intra–operative covariates associated with PPCs.

Analyses were performed using R (version 2.3; R Foundation for Statistical Computing, Vienna, Austria).

Role of funding sourceThe European Society of Anaesthesiology (ESA) and the Academic Medical Center (AMC), Amsterdam, The Netherlands financially supported and endorsed the trial. They had no influence on trial design, conduct of the trial, data analysis, or reporting.

Results

From February 2011 through January 2013, we enrolled 900 patients in 30 centres in Europe and the U.S.A. Randomization of patients was balanced within centres; 447 patients were assigned to ventilation with higher PEEP; 453 patients were assigned to ventilation with lower PEEP. Six randomized patients were excluded from analysis. Six patients receiving other treatment than allocated were kept in their original randomization arms (figure 1). Data for the primary endpoint could be analysed for 445 patients in the higher PEEP group and 449 patients in the lower PEEP group. Surgery was for cancer in 268 (61%) of the higher PEEP patients and 281 (63%) in the lower PEEP patients.

Median VT was similar between groups and remained within target throughout intra-operative mechanical ventilation. Median PEEP levels were 12 [12–12] cmH2O in the higher PEEP group and 2 [0–2] cmH2O in the lower PEEP group. The percentage of patients receiving recruitment manoeuvres following intubation was 99% in the higher PEEP group and 1% in the lower PEEP group; 85% of patients in the higher PEEP group and 0.7% in the lower PEEP group received recruitment manoeuvres before extubation (for details, see table S6). Peak pressures and dynamic respiratory compliance were significantly higher in the higher PEEP group. SpO2 levels were only marginally but statistically significantly higher in the higher PEEP group. In the higher PEEP group 2% of the patients needed a rescue for desaturation versus 8% of patients in the lower PEEP group (P < 0.0008, table 3). Details on the duration of the rescue strategy and highest step reached are described in table S7. In 34 patients in the higher PEEP group, PEEP was decreased on request of the surgeon (5 cases) or the attending anaesthesiologist (3 cases); because of hypotension (14 cases), massive surgical bleeding (10 cases), or for other reasons (2 cases).

Hemodynamic compromise and use of vasopressors occurred more frequently during the higher PEEP strategy (table 3), and these patients received more fluids (table 2). There were

PROVHILO trial

Chap

ter

8

183

Figure 2. The probability of the composite primary endpoint of postoperative pulmonarycomplications by postoperative day five between the higher PEEP group (straight line) and the lower PEEP group (dotted line) as presented by a Kaplan–Meier graph (P = 0.89, log–rank test)

Figure 1. Randomization and follow–up of study patientsNine hundred patients were randomly assigned to a study group to obtain the full sample size. Four patients withdrew informed consent for the use of their data after the end of the study intervention. One patient was randomized twice and therefore violated the exclusion criteria. One patient was randomized, but did not receive study intervention

184

Table 1. Baseline characteristics of patients

Variable Higher PEEP N = 445

Lower PEEP N = 449

Male sex – % (n/N) 58 (259/445) 57 (255/449)

Age – year, median [IQR] 65 [54 - 73] 66 [56 - 74]

BMI m2, mean (sd) 25.5 (4.2) 25.6 (4.4)

Body Weight – kg, mean (sd) 72.5 (14.3) 72.7 (14.8)

ARISCAT score – median [IQR] 41 [34 - 43] 41 [34 - 47]

Intermediate (26 - 44) – % (n/N) 78 (346/442) 74 (331/447)

High (> 44) – % (n/N) 22 (98/442) 27 (119/447)

Smoking status – % (n/N)

never 55 (245/445) 54 (242/447)

former 25 (111/445) 26 (119/449)

current 20 (91/445) 20 (91/449)

Alcohol status (past 2 weeks) – % (n/N)

none 68 (301/445) 69 (307/447)

0 - 2 units of alcohol 29 (130/445) 28 (125/447)

> 2 units of alcohol 4 (16/445) 4 (18/447)

ASA physical status classification system – % (n/N)

1 12 (55/445) 12 (54/448)

2 55 (246/445) 52 (233/448)

3 32 (142/445) 35 (156/448)

4 1 (3/445) 2 (8/448)

5 (1/445) 0

New York Heart Association Classification – % (n/N)

I 80 (347/435) 77 (339/439)

II 20 (87/435) 23 (99/439)

III 1 (3/435) 1 (4/439)

IV 0 0

Functional status – % (n/N)

non dependent 96 (427/445) 95 (426/449)

partially dependent 4 (18/445) 5 (24/449)

totally dependent 0.5 (2/445) 0.5 (2/449)

History of active cancer – % (n/N) 61 (268/441) 63 (281/448)

History of chronic renal failure – % (n/N) 6 (25/445) 5 (22/449)

COPD – % (n/N) 8 (37/445) 7 (30/449)

with inhalation therapy 3 (15/444) 3 (15/448)

with systemic steroids 2 (8/444) 2 (7/448)

PROVHILO trial

Chap

ter

8

185

Diabetes mellitus – % (n/N) 13 (56/445) 18 (79/449)

with oral medication 70 (38/54) 70 (51/73)

with insulin therapy 30 (16/54) 31 (23/74)

Use of systemic steroids – % (n/N) 2 (10/445) 2 (8/448)

Use of statins – % (n/N) 18 (82/445) 18 (80/449)

Preoperative transfusion – % (n/N) 2 (7/445) 2 (10/448)

Preoperative tests

Haemoglobin – g/L, mean (sd) 119 (26) 119 (26)

Creatinine – µmol/L, median [IQR] 61 [53 - 76] 61 [53 - 76]

Urea – mmol/L, median [IQR] 9.3 [5.7 - 13] 9.6 [5.7 - 14]

White blood cells – x109/L, median [IQR] 7 [5.7 – 8.6] 7 [5.7 – 8.7]

Pre–operative SpO2 – %, median [IQR] 97 [96 - 98] 97 [96 - 98]

Abnormalities on chest X-ray – % (n/N) 7 (23/329) 5 (18/360)

Perioperative variables

Duration of surgery† – minutes, median [IQR] 200 [140 - 300] 190 [140 - 262]

Surgical procedure – % (n/N)

gastric 9 (42/445) 9 (42/449)

pancreatic 13 (60/445) 13 (60/449)

biliary 3 (15/445) 2 (11/449)

liver 7 (31/445) 7 (31/449)

colonic 22 (100/445) 22 (98/449)

rectal 11 (50/445) 11 (48/449)

bladder 9 (39/445) 10 (47/449)

kidney 2 (10/445) 3 (12/449)

vascular 4 (16/445) 4 (18/449)

other 18 (82/445) 18 (82/449)

Antibiotic prophylaxis – % (n/N) 93 (409/440) 91 (411/449)

Type of anaesthesia – % (n/N)

total intravenous 9 (41/445) 9 (41/449)

mixed (volatile and intravenous) 91 (404/444) 91 (408/448)

Epidural – % (n/N) 49 (219/445) 50 (226/449)

thoracic 79 (173/218) 77 (174/226)

lumbar 21 (46/219) 23 (52/226)

Data is presented as: means (sd), median [IQR] or proportion % (n/N); n: number of patients; N: total patients; BMI: Body Mass Index, calculated as weight (kg)/ height (m)2 = kg/m2; kg: kilogram; m: meters; ASA: American Society of Anesthesiology; COPD: Chronic Obstructive Pulmonary Disease; Inhalation therapy for COPD: inhaled bronchodilators and/or steroids’ SpO2: oxyhaemoglobin saturation measured by pulse oximeter; †Duration of surgery is the time between skin incision and closure of the incision

186

no differences in duration of surgery, administered anaesthetics, use of epidural anaesthesia, intra–operative blood loss, transfusion of blood products, arrhythmias, surgical complications, or urine output (table 1, 2, 3, S8 and S9).

PPCs within the initial five postoperative days occurred in 174 (40%) patients in the higher PEEP group versus 172 (39%) patients in the lower PEEP group (relative risk, 1.01; 95% confidence interval 0.85–1.20; P = 0.84) (table 3 and figure 2). The need for continued or new postoperative mechanical ventilation did not differ significantly between groups: 18 (4%) patients in the higher PEEP group versus 24 (5%) patients in the lower PEEP group. Hypoxemia was relatively common at 24% in the higher PEEP group and 21% in the lower PEEP group. Discarding hypoxemia from the composite endpoint of PPCs did not result in a difference between groups (table 3). There was no heterogeneity in PPCs across centres.

In the higher PEEP group, 244 (55%) patients developed extrapulmonary complications versus 242 (54%) patients in the lower PEEP group (P = 0.78) (table 3 and figure S2). Gastro–intestinal failure was the most common extrapulmonary complication, followed by SIRS and acute renal failure, but distributed equally between randomization groups (table 3).

There was no difference in the need for intensive care unit admission, number of hospital free days at postoperative day 90, nor in hospital mortality (table 3).

The results of a per–protocol analysis were not different from the intention–to–treat analysis (table S10). The results of the post–hoc analysis of the association between the incidence of postoperative pulmonary complications and intra–operative use of medication (anaesthetics, neuromuscular blocking agents, and opioids), and a multiple logistic regression analysis to identify baseline and intra–operative covariates, which are associated with PPCs are shown in the Supplementary Appendix (tables S9 and S11).

Discussion

PROVHILO is the first trial in which identical lower VTs were used in both study arms, making it possible to isolate the effects of higher PEEP levels from the known effects of VT size. In 900 patients at risk for PPCs after mechanical ventilation under general anaesthesia for open abdominal surgery, the incidence of PPCs within the first five postoperative days was comparable between patients receiving higher PEEP with recruitment manoeuvres and lower PEEP without recruitment manoeuvres.

Our composite endpoint of PPCs included hypoxemia, which was the most common PPC. Restricting our analysis to more severe PPCs did not change the study results, suggesting that PEEP level does not alter the risk of more severe pulmonary complications. The incidence of PPCs in the present trial was substantially higher than in these previous investigations,1,3,9,15,16 which could have been caused by the much higher risk of developing PPC’s as compared to patients in previous studies. Because the observed incidence was so high, our trial had sufficient statistical

PROVHILO trial

Chap

ter

8

187

Table 2. Intraoperative ventilation characteristics

Variable Higher PEEP N = 445

Lower PEEP N = 449 p-value

Tidal volumes – mL, median [IQR] 500 [450 – 560] 500 [450 – 550]

Tidal volumes – mL/kg PBW, mean (sd) 7.2 (1.5) 7.1 (1.2)

Tidal volumes, after 1 hour 7.11 (1.32) 7.09 (1.23)

Tidal volumes, directly before extubation 6.96 (1.50) 7.07 (1.23)

PEEP – cmH2O, median [IQR] 12 [12 – 12] 2 [0 – 2]

PEEP, after 1 hour 12 [12 – 12] 2 [0 – 2]

PEEP, directly before extubation 12 [12 – 12] 2 [0 – 2]

Peak pressure – mL/cmH2O, mean (sd) 23 (3.7) 17 (4.1)

Peak pressure – mL/cmH2O, after 1 hour 23.1 (4.1) 16.8 (4.4)

Peak pressure – mL/cmH2O, directly before extubation 22.7 (4.2) 16.7 (4.1)

Cdyn (calculated) – cmH2O, median [IQR] 44 [35 – 54] 34 [27 – 41] < 0.0001

Cdyn, begin# 45 [36 – 57] 33 [27 – 43] < 0.0001

Cdyn, end# 44 [36 – 54] 35 [27 – 42] < 0.0001

Respiratory rate – breaths/min, mean (sd) 11 (2.1) 11 (1.9) 0.13

Minute ventilation – mL/min, mean (sd) 5681 (1267) 5545 (1162) 0.10

FiO2 – %, median [IQR] 40 [40 – 49] 41 [40 – 50] 0.06

< 40% – % (n/N)* 50 (222/445) 45 (202/449) 0.14

40 – 60 43 (190/445) 46 (206/449) 0.34

60 – 80 4 (18/445) 5 (22/449) 0.54

> 80% 3 (15/445) 4 (19/449) 0.50

SpO2 – %, median [IQR] 99 [98.5 – 100] 99 [98 – 99.8] < 0.0001

FE’CO2 – mmHg, mean (sd) 35.2 (3.7) 34.5 (3.4) < 0.0007

BP mean – mmHg, mean (sd) 77.8 (9.8) 77.9 (10) 0.28

> 70 – % (n/N)* 61 (270/445) 60 (269/449) 0.82

60 – 70 31 (137/445) 30 (134/449) 0.76

< 60 9 (38/445) 10 (46/449) 0.38

HR – bpm, mean (sd) 70.7 (12.7) 68.8 (10.9) 0.0121

Patients receiving RM after intubation – % (n/N) 99 (438/442) 1 (6/452)

188

power to detect a difference in the incidence of PPCs of 7.5%. We made efforts to minimize the risk of bias through centralized randomization, and blinding of study group assignment for outcome assessors. We used a relevant composite outcome at a meaningful interval in this surgical population. In addition, we published the statistical analysis plan before unblinding study group assignments.14

The chosen PEEP level in the higher PEEP group is supported by the literature.17,18 Previous studies tested PEEP levels of 10 cmH2O during intra–operative ventilation,19-21 but atelectasis persisted during anaesthesia in some patients, especially when higher FIO2 was used.21 Notably, these atelectasis may also persist in the first postoperative days, especially after abdominal surgery.22

Variable Higher PEEP N = 445

Lower PEEP N = 449 p-value

Patients receiving RM before extubation – % (n/N) 85 (378/444) 0.7 (3/429)

Crystalloids, median [IQR] 2200 2000 0.0229

[1500 - 3100] [1400 - 3000]

Colloids, median [IQR] 500 [0 - 1000] 500 [0 - 1000] 0.30

Total fluids – % (n/N)$

< 1000 mL 5 (22/436) 9 (41/435) 0.0126

1000 – 3000 mL 54 (236/436) 56 (245/435) 0.52

3000 – 5000 mL 30 (131/436) 26 (111/435) 0.14

> 5000 mL 11 (47/436) 9 (38/435) 0.31

Urine output – mL, median [IQR] 300 [187 - 560] 340 [200 - 600] 0.32

PRBC transfused – % (n/N) 14 (62/443) 17 (78/ 449) 0.24

FFP transfused – % (n/N) 5 (21/420) 5 (24/449) 0.82

Platelets transfused – % (n/N) 1 (3/429) 2 (10/449) 0.0559

Blood loss – mL, median [IQR] 500 [200 - 1000] 400 [200 - 800] 0.38

Massive transfusion – % (n/N) 2.7 (12/444) 1.1 (5/445) 0.09

Temperature at end of surgery – ⁰C, mean (sd) 36 (0.6) 36 (0.6) 0.58

Perforation organ – % (n/N) 0.9 (4/444) 0.9 (4/444) 1

Data is presented as means (sd); median [IQR] or proportion % (n/N); n: number of patients; N: total patients; PBW: predicted body weight, calculated as: 50 + 0.91 x (centimetres of height – 152.4) for males and 45.5 + 0.91 x (centimetres of height – 152.4) for females; #Begin: during the first hour of mechanical ventilation; End: during the last hour before extubation; PEEP: positive end-expiratory pressure; Cdyn: calculated dynamic respiratory compliance, calculated as VT/ (Ppeak – PEEP) = mL/cmH2O; Ppeak: peak pressure; FiO2: fraction inspired oxygen; SpO2: Oxyhaemoglobin saturation measured by pulse oximeter; FE’CO2: expiratory carbon dioxide partial pressure; BP: blood pressure; HR: heart rate; bpm: beats per minute; RM: recruitment manoeuvre; PRBC: packed red blood cells; FFP: fresh frozen plasma; ⁰C = degrees Celsius; *Categories of FiO2 and BPmean are scored upon occurrence of worst clinical parameter (no., %) $Total fluids are crystalloids and colloids

PROVHILO trial

Chap

ter

8

189

Table 3. Primary and Secondary Outcomes

Variable Higher PEEP N = 445

Lower PEEP N = 449 p-value RR (95% CI)

Postoperative Pulmonary Complications* – % (n/N)

Combined PPCs 40 (174/437) 39 (172/443) 0.84 1.01 (0.85 – 1.20)

Combined PPCs (excluding hypoxemia) 32 (142/437) 34 (149/443) 0.66 0.96 (0.78 – 1.17)

Hypoxemia 24 (105/437) 21 (95/443) 0.36 1.08 (0.92 – 1.25)

Severe hypoxemia 7 (29/437) 8 (34/443) 0.55 0.92 (0.70 – 1.21)

Bronchospasm 4 (18/437) 4 (18/443) 0.97 1.01 (0.72 – 1.41)

Suspected pulmonary infection 16 (68/437) 17 (75/443) 0.58 0.95 (0.79 – 1.14)

Pulmonary infiltrate 8 (35/437) 7 (32/443) 0.66 1.06 (0.83 – 1.34)

Aspiration pneumonitis 0.2 (1/437) 1 (4/443) 0.18 0.40 (0.07 – 2.32)

ARDS 1 (5/437) 2 (8/443) 0.41 0.77 (0.39 – 1.54)

Atelectasis 12 (53/437) 12 (55/443) 0.90 0.99 (0.80 – 1.21)

Pleural effusion 21 (90/437) 21 (92/443) 0.95 0.99 (0.84 – 1.17)

Pulmonary oedema caused by cardiac failure 4.3 (19/437) 4.5 (20/443) 0.90 0.98 (0.71 – 1.36)

Pneumothorax 3.4 (15/437) 2.7 (12/443) 0.53 1.12 (0.80 – 1.58)

Postoperative Extrapulmonary Complications* – % (n/N)

Combined extrapulmonary complications 55 (244/445) 54 (242/449) 0.78 1.02 (0.90 – 1.15)

SIRS 14 (62/437) 14 (64/443) 0.91 0.97 (0.70 – 1.35)

Sepsis 4 (18/437) 4 (18/443) 0.96 1.01 (0.53 – 1.91)

Severe sepsis 1 (5/437) 1 (4/443) 0.72 1.26 (0.34 – 4.67)

Septic shock 1 (3/437) 1 (3/443) 0.98 1.01 (0.20 – 4.97)

Extrapulmonary infections 8 (34/437) 7 (31/443) 0.66 1.11 (0.69 – 1.77)

Coma 0 (1/437) 0 (1/443) 0.49 1.01 (0.06 – 16)

Acute myocardial infarction 1 (6/437) 1 (5/443) 0.74 1.21 (0.37 – 3.94)

Acute renal failure (RIFLE criteria)** 0.60

No 87 (342/391) 86 (341/397) 0.52 1.02 (0.96 – 1.08)

Risk 9 (34/391) 8 (33/397) 0.85 1.05 (0.66 – 1.65)

Injury 2 (8/391) 4 (14/397) 0.21 0.58 (0.25 – 1.37)

Failure 2 (7/391) 2 (9/397) 0.64 0.79 (0.30 – 2.10)

Loss 0.2 (1/391)

190

Variable Higher PEEP N = 445

Lower PEEP N = 449 p-value RR (95% CI)

Disseminated intravascular coagulation 0.2 (1/437) 0 0.16 0.14 (0.02 –1.17)

Hepatic Failure 7 (32/445) 8 (34/449) 0.84 0.95 (0.60 – 1.52)

Gastro-intestinal bleeding 1 (3/ 437) 1 (6/443) 0.32 0.51 (0.13 – 2.03)

Gastro-intestinal failure** 0.94

Score 0 50 (197/394) 48 (193/399) 0.79 1.03 (0.89 – 1.20)

Score 1 41 (162/394) 42 (168/399) 0.86 0.98 (0.82 – 1.18)

Score 2 8 (33/394) 9 (35/399) 0.85 0.96 (0.61 – 1.51)

Score 3 0.5 (2/394) 1 (3/399) 0.66 0.68 (0.11 – 4.03)

Score 4 0 0

Intra-operative complications – % (n/N)Ŧ

Rescue strategy for de–saturation 2 (11/442) 8 (34/445) < 0.0008 0.34 (0.18 – 0.67)

Hypotension 46 (205/441) 36 (162/449) 0.0016 1.29 (1.10 – 1.51)

Vasoactive drugs 62 (274/444) 51 (228/445) 0.0016 1.20 (1.07 – 1.35)

New arrhythmias 3 (12/442) 1 (5/445) 0.09 2.38 (0.84 – 6.70)

Follow–up

Impaired wound healing – % (n/N) 16 (71/444) 13 (58/446) 0.21 1.23 (0.89 – 1.70)

Need for new or continued MV – % (n/N) 4 (18/437) 5 (24/443) 0.74 0.77 (0.42 – 1.40)

ICU admission – % (n/N) 24 (106/442) 23 (104/452) 0.79 1.03 (0.81 – 1.32)

Length of hospital stay – days, median [IQR] 10 [7 – 14] 10 [7 – 14] 0.24 1.01 (0.42 – 2.40)

Hospital free days at day 90, median [IQR] 79 [71 – 83] 79 [70 – 82] 0.33

Mortality by day 5 – % (n/N) 0.4 (2/443) 0.2 (1/448) 0.56 2.02 (0.18 – 22)

In hospital mortality – % (n/N) 2 (7/ 438) 2 (7/442) 0.99 1.01 (0.36 – 2.85)

Data is presented as means ± (sd), median [IQR] or proportion % (n/N); n: number of patients; N: total patients and relative risk with 95% Confidence Intervals; PPCs: postoperative pulmonary complications; ARDS: acute respiratory distress syndrome; SIRS: systemic inflammatory response syndrome; Renal failure documented as follows: Risk: increased creatinine x1.5 or glomerular filtration rate (GFR) decrease > 25% or urine output (UO) < 0.5 ml/kg/h x 6 h; Injury: increased creatinine x2 or GFR decrease > 50% or UO < 0.5 ml/kg/h x 12 hr; Failure: increase creatinine x3 or GFR decrease > 75% or UO < 0.3 ml/kg/h x 24 hr or anuria x 12 hrs; Loss: persistent ARF = complete loss of kidney function > 4 weeks; Gastro-intestinal failure score: 0 = normal gastrointestinal function; 1 = enteral feeding with under 50% of calculated needs or no feeding 3 days after abdominal surgery; 2 = food intolerance (FI) or intra–abdominal hypertension (IAH); 3 = FI and IAH; and 4 = abdominal compartment syndrome; Impaired wound healing is defined as an interruption in the timely and predictable recovery of mechanical integrity in the injured tissue; MV: mechanical ventilation; ICU: intensive care unit*Pulmonary complications, and extrapulmonary complications and impaired wound healing on day 1 to 5 were scored YES as soon as an event occurred; **Acute Renal failure & Gastro-intestinal failure; highest value occurring in day 1 to 5 is scored; ŦIntra-operative complications were scored YES as soon as complication occurred; Rescue strategy for desaturation (SpO2 < 90%) performed as described in methods; Hypotension defined as systolic arterial blood pressure < 90 mmHg for more than 3 minutes; Vasoactive drugs defined as need for vasoactive medication; New arrhythmias requiring intervention

PROVHILO trial

Chap

ter

8

191

We chose a PEEP level of 12 cmH2O to maximize lung opening throughout mechanical ventilation, irrespective of FIO2. The higher PEEP strategy resulted in improved dynamic compliance of the respiratory system compared to the lower PEEP group, suggesting higher alveolar recruitment.

The results of the present trial expand the understanding of findings of two recently published trials comparing a conventional ventilation strategy with higher VTs and no PEEP with a protective strategy using lower VTs and higher levels of PEEP in similar patient populations.9,10 It is suggested that benefit of protective ventilation in those trials must have come from the higher levels of PEEP.23 However, the design of those trials does not allow identifying whether lower VTs, higher levels of PEEP, or both, are responsible for the beneficial effects. The results of the present trial challenge the hypothesis that higher PEEP is responsible for the beneficial effects of protective ventilation. However, it must be kept in mind that trials are not completely comparable, as the levels of higher PEEP in the previous trials9,10 were approximately 4 – 6 cmH2O lower than those in the present trial.

Perhaps in our trial, higher PEEP stabilized the lungs and protected against lung injury from tidal recruitment, but the adverse effects counteracted the possible beneficial effects. Peak airway pressures were increased in the higher PEEP group, possibly causing hyperinflation in non–dependent lung zones. Furthermore, higher PEEP further impaired the hemodynamics. Thus, our findings suggest that levels of PEEP higher than recommended in previous trials,9,10 although improving the elastic properties of the respiratory system does not enhance lung protection in general anaesthesia.

Several drugs used for general anaesthesia induce peripheral vascular smooth muscle relaxation, decrease the arterial pressure, and even impair cardiac contractility.24,25 Also, epidural anaesthesia, which is frequently used in combination with general anaesthesia during open abdominal surgery in up to 50% of cases, may contribute to reduce the peripheral vascular smooth muscle tonus and promote peripheral blood pooling.26 However, there was neither difference in the administration of drugs for general anaesthesia, nor in use of epidural anaesthesia between the groups. The increased incidence of intra–operative hemodynamic adverse events in the higher PEEP group, especially arterial hypotension, thus may have been associated with a reduction of the venous return due to increased intrathoracic with higher PEEP and/or recruitment manoeuvres. Even though those events were limited and responded to increased intravascular volume expansion, as well as use of vasoactive drugs, they might be threatening in presence of ischemic cardiac disease.27

We did not include patients having laparoscopic surgery or morbidly obese patients, both groups that may have especially benefited from higher intra–operative PEEP. Furthermore, we recommended but did not reinforce use of international guidelines and standards for intra–operative and postoperative fluid administration, use of inotropes and/or vasopressors, and use and/or reversal of neuromuscular blocking agents. Our study was pragmatic in its design, rather than tightly controlled. As randomization was balanced within the centres, thus we consider it unlikely that this affected the trial results. As randomization was balanced within the centres, it is unlikely to have affected our results. A corollary is that our results are relatively generalizable to a broad range of practice styles. A corollary is that our results are relatively generalizable

192

to a broad range of practice styles. The use of an equally weighed composite endpoint could be seen as a limitation as well, but we have given insight into the distribution of the events by presenting the incidence of each complication separately.

In conclusion, during mechanical ventilation with protective lower VT in patients undergoing open abdominal surgery, the use of higher levels of PEEP and recruitment manoeuvres does not reduce the incidence of PPCs, and more frequently results in hemodynamic instability, as compared to lower PEEP without recruitment manoeuvres.

Panel: Research in context

Systematic reviewApproximately 234 million major surgical procedures are performed worldwide every year, with major impact on the global economy. Among these interventions, around 2.6 million represent high–risk procedures, with 1.3 million patients developing complications that result in 315,000 in–hospital deaths.28 Postoperative pulmonary complications (PPCs) are frequent during non-cardiac surgery and associated with increased risk of in-hospital death, especially after open abdominal surgery.2,3 Notably, the ventilatory strategy applied to these patients may impact outcomes. A recent metaanalysis showed namely that lower tidal volume ventilation is associated with reduced pulmonary and extrapulmonary complications, as well as lower mortality in patients without previous lung injury.4 Also, another metaanalysis showed that mechanical ventilation during general anaesthesia with lower tidal volume and higher positive end-expiratory pressure (PEEP) plus recruitment manoeuvres are associated with lower incidence of PPCs and improved respiratory function when compared to higher tidal volumes and lower PEEP.29 Two recently published trials showed that using lower tidal volumes and higher levels of PEEP with recruitment manoeuvres prevents PPCs and reduces healthcare resources in abdominal surgery.9,10 Nevertheless, clinical evidence so far does not allow determining whether intra-operative use of higher levels of PEEP or use of lower tidal volumes or both are responsible for protection against PPCs. We thus investigated the potential of higher PEEP with recruitment manoeuvres versus lower PEEP to protect against PPCs in 900 patients undergoing general anaesthesia for open abdominal surgery and mechanical ventilation with low tidal volume.

InterpretationAs far as we are aware, this is the largest multicentre, international, randomized controlled trial of mechanical ventilation during general anaesthesia for open abdominal surgery investigating the isolated role of higher PEEP plus recruitment manoeuvres against PPCs. We found that a strategy using higher levels of PEEP and recruitment manoeuvres as compared to lower levels of PEEP without recruitment manoeuvres does not reduce the incidence of PPCs, while increasing intra-operative circulatory impairment. These findings may change current practice of mechanical ventilation during general anaesthesia for open abdominal surgery.

PROVHILO trial

Chap

ter

8

193

References1. Smetana GW, Lawrence VA, Cornell JE. Preoperative pulmonary risk stratification for noncardiothoracic surgery:

systematic review for the American College of Physicians. Annals of internal medicine 2006; 144: 581-952. Pearse RM, Moreno RP, Bauer P, Pelosi P, Metnitz P, Spies C, et al. Mortality after surgery in Europe: a 7 day cohort

study. Lancet 2012; 380: 1059-653. Canet J, Gallart L, Gomar C, Paluzie G, Valles J, Castillo J, et al. Prediction of Postoperative Pulmonary Complications

in a Population-based Surgical Cohort. Anesthesiology 2010; 113: 1338-504. Serpa Neto A, Cardoso SO, Manetta JA, Pereira VG, Esposito DC, Pasqualucci Mde O, et al. Association between use of

lung-protective ventilation with lower tidal volumes and clinical outcomes among patients without acute respiratory distress syndrome: a meta-analysis. JAMA 2012; 308: 1651-9

5. Lellouche F, Dionne S, Simard S, Bussieres J, Dagenais F. High tidal volumes in mechanically ventilated patients increase organ dysfunction after cardiac surgery. Anesthesiology 2012; 116: 1072-82

6. Duggan M, Kavanagh BP. Pulmonary atelectasis: a pathogenic perioperative entity. Anesthesiology 2005; 102: 838-547. Putensen C, Theuerkauf N, Zinserling J, Wrigge H, Pelosi P. Metaanalysis: ventilation strategies and outcomes of the

acute respiratory distress syndrome and acute lung injury. Annals of internal medicine 2009; 151: 566-768. Briel M, Meade M, Mercat A, Brower RG, Talmor D, Walter SD, et al. Higher vs lower positive end-expiratory pressure

in patients with acute lung injury and acute respiratory distress syndrome: systematic review and metaanalysis. JAMA 2010; 303: 865-73

9. Severgnini P, Selmo G, Lanza C, Chiesa A, Frigerio A, Bacuzzi A, et al. Protective Mechanical Ventilation during General Anesthesia for Open Abdominal Surgery Improves Postoperative Pulmonary Function. Anesthesiology 2013; 118: 1307-21

10. Futier E, Constantin JM, Paugam-Burtz C, Pascal J, Eurin M, Neuschwander A, et al. A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. NEJM 2013; 369: 428-37

11. Halter JM, Steinberg JM, Schiller HJ, DaSilva M, Gatto LA, Landas S, et al. Positive end-expiratory pressure after a recruitment maneuver prevents both alveolar collapse and recruitment/derecruitment. American journal of respiratory and critical care medicine 2003; 167: 1620-6

12. Nielsen J, Ostergaard M, Kjaergaard J, Tingleff J, Berthelsen PG, Nygard E, et al. Lung recruitment maneuver depresses central hemodynamics in patients following cardiac surgery. Intensive Care Medicine 2005; 31: 1189-94

13. Carvalho AR, Pacheco SA, de Souza Rocha PV, Bergamini BC, Paula LF, Jandre FC, et al. Detection of tidal recruitment/overdistension in lung-healthy mechanically ventilated patients under general anesthesia. Anesthesia and Analgesia 2013; 116: 677-84

14. Hemmes SN, Severgnini P, Jaber S, Canet J, Wrigge H, Hiesmayr M, et al. Rationale and study design of PROVHILO - a worldwide multicenter randomized controlled trial on protective ventilation during general anesthesia for open abdominal surgery. Trials 2011; 12: 111

15. Arozullah AM, Daley J, Henderson WG, Khuri SF. Multifactorial risk index for predicting postoperative respiratory failure in men after major noncardiac surgery. The National Veterans Administration Surgical Quality Improvement Program. Ann Surg 2000; 232: 242-53

16. Arozullah AM, Khuri SF, Henderson WG, Daley J. Development and validation of a multifactorial risk index for predicting postoperative pneumonia after major noncardiac surgery. Annals of Internal Medicine 2001; 135: 847-57

17. Tusman G, Bohm SH, Vazquez de Anda GF, do Campo JL, Lachmann B. ‘Alveolar recruitment strategy’ improves arterial oxygenation during general anaesthesia. British journal of anaesthesia 1999; 82: 8-13

18. Tusman G, Belda JF. Treatment of anesthesia-induced lung collapse with lung recruitment maneuvers. Current Anaesthesia & Critical Care 2010; 21: 244-9

19. Brismar B, Hedenstierna G, Lundquist H, Strandberg A, Svensson L, Tokics L. Pulmonary densities during anesthesia with muscular relaxation - a proposal of atelectasis. Anesthesiology 1985; 62: 422-8

20. Hachenberg T, Lundquist H, Tokics L, Brismar B, Hedenstierna G. Analysis of lung density by computed tomography before and during general anaesthesia. Acta Anaesthesiologica Scandinavica 1993; 37: 549-55

21. Neumann P, Rothen HU, Berglund JE, Valtysson J, Magnusson A, Hedenstierna G. Positive end-expiratory pressure prevents atelectasis during general anaesthesia even in the presence of a high inspired oxygen concentration. Acta Anaesthesiologica Scandinavica 1999; 43: 295-301

22. Lindberg P, Gunnarsson L, Tokics L, Secher E, Lundquist H, Brismar B, et al. Atelectasis and lung function in the postoperative period. Acta Anaesthesiologica Scandinavica 1992; 36: 546-53

23. Melo MF, Eikermann M. Protect the lungs during abdominal surgery: it may change the postoperative outcome. Anesthesiology 2013; 118: 1254-7

24. Pagel P, Farber N, Pratt P, Warltier D. Cardiovascular Pharmacology. In: Miller R, Eriksson L, Fleisher L, Wiener-Kronish J, Young W, ed. Miller’s Anesthesia. 7th ed. Philadelphia, PA: Elsevier, 2009: 595-632

194

25. Reves J, Glass P, Lubarsky D, McEvoy M, Martinez-Ruiz R. Intravenous Anesthetics. In: Miller R, Eriksson L, Fleisher L, Wiener-Kronish J, Young W, ed. Miller’s Anesthesia. 7th ed. Philadelphia, PA, USA: Elsevier, 2009: 719-68

26. Brown D. Spinal, Epidural and Caudal Anesthesia. In: Miller R, Eriksson L, Fleisher L, Wiener-Kronish J, Young W, ed. Miller’s Anesthesia. 7th ed. Philadelphia, PA, USA: Elsevier, 2009: 1611-38

27. Luecke T, Pelosi P. Clinical review: Positive end-expiratory pressure and cardiac output. Critical Care 2005; 9: 607-2128. Weiser TG, Regenbogen SE, Thompson KD, Haynes AB, Lipsitz SR, Berry WR, et al. An estimation of the global volume

of surgery: a modelling strategy based on available data. Lancet 2008; 372: 139-4429. Hemmes SN, Serpa Neto A, Schultz MJ. Intraoperative ventilatory strategies to prevent postoperative pulmonary

complications: a metaanalysis. Current Opinion in Anaesthesiology 2013; 26: 126-33

PROVHILO trial

Chap

ter

8

195

Supplementary Appendix to ‘Higher versus lower positive end-expiratory pressure during general anaesthesia for open abdominal surgery – The PROVHILO trial’

List of PROVE Network InvestigatorsWriting committeeSabrine N.T. Hemmes (Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands); Marcelo Gama de Abreu (University Hospital Dresden, Germany); Paolo Pelosi (University of Genoa, Genoa, Italy); Marcus J. Schultz (Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands)

Steering committeeSabrine N.T. Hemmes (Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands); Paolo Severgnini (University of Insubria, Varese, Italy); Samir Jaber (Saint Eloi University Hospital, Montpellier, France); Jaume Canet (Hospital Universitari Germans Trias I Pujol, Barcelona, Spain); Hermann Wrigge (University of Leipzig, Germany); Michael Hiesmayr (Medical University, Vienna, Austria); Werner Schmid (Medical University, Vienna, Austria); Markus W. Hollmann (Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands); Jan M. Binnekade (Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands); Göran Hedenstierna (University Hospital, Uppsala, Sweden); Christian Putensen (University Hospital, Bonn, Germany); Marcelo Gama de Abreu (University Hospital Dresden, Germany); Paolo Pelosi (University of Genoa, Genoa, Italy); Marcus J. Schultz (Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands)

StatisticianJan M. Binnekade (Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands)Data safety and monitoring boardDaniel I. Sessler (chair) (Michael Cudahy Professor & Chair, Department of Outcomes Research, Cleveland Clinic, Cleveland, U.S.A.), Burkhard Lachmann (Professor emeritus, Department of Anaesthesia and Intensive Care Medicine at the Universitätsmedizin Berlin, Charite; Campus Virchow Klinikum, Germany), Robert M. Kacmarek (Professor of Anesthesiology, Harvard Medical School and Director Respiratory Care, Massachusetts General Hospital, Boston, MA, U.S.A.) and Arthur S. Slutsky (Professor at St. Michaels Hospital, University of Toronto, Toronto, ON, Canada and Keenan Research Center of the Li Ka Shing Institute of St. Michael’s Hospital, Toronto)

PROVE Network websitewww.provenet.eu

PROVE Network Collaborators (*, indicates local principal investigator; names are listed in alphabetical order)

AustriaMedical University Vienna: Werner Schmid (national coordinator)*

BelgiumGhent University Hospital: Luc De Baerdemaeker, Stefan De Hert (national coordinator)*, Bjorn Heyse, Jurgen Van LimmenAZ St Jan, Brugge: Jan–Paul Mulier*ZNA Middelheim, Antwerpen: David Velghe*Virga Jesse Ziekenhuis, Hasselt: Luc Jamaer*, Jeroen Vandenbrande Republic of Chile Hospital Clínico de la Pontificia Universidad Católica de Chile, Santiago: Guillermo Bugedo (national coordinator)*, Jorge Florez

CroatiaUniversity Hospital Sveti Duh, Zagreb: Tatjana Goranović, Branka Mazul–Sunko (national coordinator)*

196

GermanyUniversity Hospital Dresden: Thomas Bluth, Marcelo Gama de Abreu (national coordinator)*, Andreas Güldner, Thomas Kiss, Thea Koch, Peter Markus Spieth, Christopher Uhlig, Jonathan YaqubDüsseldorf University Hospital, Heinrich-Heine University Düsseldorf: Bea Bastin, Johann Geib, Maximilian S. Schaefer, Martin Weiss, Tanja A. Treschan*University of Leipzig: Andreas W. Reske, Philipp Simon, Hermann Wrigge* Johannes Gutenberg - Universität Mainz: Alexander Brodhun Marion Ferner, Eric Hartmann, Rita Laufenberg-Feldmann*, Lydia StrysUniversity Hospital of Bonn Medical School: Christian Putensen*

ItalyUniversity of Napoli Federico II, Naples: Edoardo De Robertis (national coordinator)* Università degli Studi di Roma Cattolica: Valter Perilli, Rodolfo Proietti*University “Magna Graecia” of Catanzaro: Bruno Amantea, Santo Caroleo*, Francesco TropeaUniversity of Insubria - Azienda Ospedaliera Fondazione Macchi - Ospedale di Circolo - Varese: Alessandro Bacuzzi, Paolo Severgnini*, Massimo VanoniUniversity of Foggia: Gilda Cinnella*, Girolamo Caggianelli, Davide D’Antini, Daniela La Bella, Giuseppina MollicaUniversità degli Studi di Palermo: Andrea Cortegiani, Antonino Giarratano*, Francesca Montalto, Santi Maurizio RaineriAzienda Sanitaria Locale TO3 - Ospedale di Rivoli, Torino: Bruno Barberis, Cristian Celentano, Michele Grio, Luigi Spagnolo*Università degli Studi di Genova: Angelo Gratarola, Alexandre Molin*, Giulia Pellerano, Stefano Pezzato, Roberta Rusca Università degli Studi di Udine: Giorgio Della Rocca*

The NetherlandsAcademic Medical Center, University of Amsterdam: Lieuwe D.J. Bos, Sabrine N.T. Hemmes (national coordinator), Markus W. Hollmann, Marcus J. Schultz*

SpainHospital Universitari Germans Trias I Pujol, Barcelona: Andrea Brunelli*, Agnes MartiHospital Sant Pau, Barcelona: Virginia Cegarra, Alfred Merten, Mª Victoria Moral, Ana Parera, Mª Carmen Unzueta*Fundación Puigvert, Barcelona: Sergi Sabate*, Pilar Sierra, Juan F Mayoral, Mercè PrietoConsorcio Hospital General Universitario Valencia: Manuel Granell Gil*, Conrado Minguez Marín

United KingdomSheffield Teaching Hospitals: Gary H. Mills (national coordinator)*Barts Health NHS Trust, London: Phoebe Bodger*

United States of AmericaMassachusetts General Hospital, Boston: Marcos F. Vidal Melo (national coordinator)*, Demet SulemanjiMayo Clinic, Rochester, Minnesota: Juraj Sprung*

AcknowledgementsWe are indebted to all participating research nurses, nurse anaesthetists, surgeons, other physicians and our patients. Without them the PROVHILO trial would never have been successful. We also thank Brigitte Leva and Sandrine Damster from the Research Team at the European Society of Anaesthesiology for their help and Annelou van der Veen for performing the on-site monitoring. We are particularly grateful to Prof. Daniel I. Sessler for revising the manuscript

List of supporting investigatorsAnn De Bruyne (Ghent University Hospital, Belgium), Patricia Ongena (ZNA Middelheim, Antwerpen, Belgium), Jörg-Uwe Bleyl, Moritz Koch, Michael Müller, Thomas Rössel, Hans-Detlef Saeger, Jürgen Weitz (University Hospital Dresden, Germany), Renate Babian, Anna Malina Rathmann (Heinrich-Heine-University Düsseldorf, Germany), Julia Pochert, Mandy Dathe (University of Leipzig, Germany), Fernando Chiaravalloti, Daniela Madia, Ivana Pezzoli, Andrea Caruso, Maria Francesca Bianco, Francesco Picicco (University “Magna Graecia” of Catanzaro, Italy), Lucia Mirabella, Michela Rauseo, Romina Anguilano (University of Foggia, Italy), Cesira Palmeri, Maria Teresa Strano, Antonino Federico (Università degli Studi di Palermo, Italy), Livia Pompei, Stefania Buttera (Università degli Studi di Udine, Italy), Kirsty Everingham, Ruth Han, Russell Hewson, Marta Januszewska, Otto Mohr, Rupert Pearse, Ashok Raj (Barts Health NHS Trust, London, U.K.), Jun Oto, Robert M. Kacmarek (Massachusetts General Hospital, Boston, U.S.A.), Toby N. Weingarten (Mayo Clinic, Rochester, Minnesota, U.S.A.)

PROVHILO trial

Chap

ter

8

197

FundingThe European Society of Anaesthesiology (ESA) financially supported and endorsed the trial. The Academic Medical Center (AMC), Amsterdam, The Netherlands sponsored the trial. Neither the ESA nor the AMC had influence on trial design of the trial, conduct of the trial, data analysis, or reporting

Analysis treatment group (high peep) on the postoperative pulmonary complications, controlling for centerWe analysed the net effect of the treatment group (high peep) on the postoperative pulmonary complications, controlling for center. Therefore we explored the interaction (effect modification) and confounding. We first focussed on the crude (uncorrected) effect of the treatment group (independent variable) on postoperative pulmonary complications (dependent variable): Odds Ratio 1.04 (95% CI 0.80 - 1.37), p 0.76. Then the variable ‘center’ was added as an interaction term. If the interaction terms appeared to be significant (p<0.05), this would indicate that the relation between the treatment group and pulmonary complications could be different for various levels of the covariate (the centers). This would indicate the need for separate models to explore the levels of the covariate. A significant interaction was not found, so the model was examined for confounding: Odds Ratio 1.05 (95% CI 0.78 - 1.42). This concludes that there is no indication that heterogeneity between participating centers is of any significant influence. In conclusion ‘center’ as covariate is not an interaction term or a confounding term on the treatment effect: postoperative pulmonary complications

Figure S1. Kaplan-Meier curve for postoperative pulmonary complications: all patientsThe probability of the composite primary endpoint of postoperative pulmonary complications by postoperative day five as presented by a Kaplan–Meier and the 95% confidence interval

198

Figure S2. Kaplan-Meier curve for extrapulmonary complications The probability of the composite endpoint of extrapulmonary complications by postoperative day five between the higher PEEP group (straight line) and the lower PEEP group (dotted line) as presented by a Kaplan–Meier graph (P = 0.83, log–rank test)

PROVHILO trial

Chap

ter

8

199

Table S1. Inclusion and exclusion criteria

Inclusion criteria

Planned open abdominal surgery

General anaesthesia

High or intermediate risk for postoperative pulmonary complications following abdominal surgery, according to the ARISCAT risk score1 (higher or equal than 26) (table S2)

Exclusion criteria

Age < 18 years

Body mass index > 40 kg/m2

Laparoscopic surgery

Previous lung surgery (any)

Persistent hemodynamic instability, intractable shock (considered hemodynamic unsuitable for the study by the patient’s managing physician)

History of previous severe chronic obstructive pulmonary disease (COPD); non–invasive ventilation, and/or oxygen therapy at home or repeated systemic corticosteroid therapy for acute exacerbations of COPD

Recent immunosuppressive medication (receiving chemotherapy or radiation therapy within last 2 months)

Severe cardiac disease (New York Heart Association class III or IV, or acute coronary syndrome, or persistent ventricular tachyarrhythmia’s)

Mechanical ventilation > than 30 minutes (e.g., in cases of general anaesthesia because of surgery) within last 30 days

Pregnancy (excluded by laboratory analysis)

Acute lung injury or acute respiratory distress syndrome expected to require prolonged postoperative mechanical ventilation

Neuromuscular disease (any)

Consented for another interventional study or refusal to participate in the study

200

Table S2. ARISCAT risk score Independent predictors of risk for post-operative pulmonary complications identified in the logistic regression model

Multivariate ModelOR (95% CI)n = 1,624

β Coefficient Risk Score†

Age (years)

≤ 50 1

51 – 80 1.4 (0.6 – 3.3) 0.331 3

> 80 5.1 (1.9 – 13.3) 1.619 16

Pre-operative (SpO2, %)

≥ 96 1

91 – 95 2.2 (1.2 – 4.2) 0.802 8

≤ 90 10.7 (4.1 – 28.1) 2.375 24

Respiratory infection 5.5 (2.6 – 11.5) 1.698 17

in the last month

Pre-operative anaemia (≤ 10 g/dl) 3.0 (1.4 – 6.5) 1.105 11

Surgical incision

Peripheral 1

Upper abdominal 4.4 (2.3 – 8.5) 1.480 15

Intra-thoracic 11.4 (4.9 – 26.0) 2.431 24

Duration of surgery (hours)

≤ 2 1

2 – 3 4.9 (2.4 – 10.1) 1.593 16

> 3 9.7 (4.7 – 19.9) 2.268 23

Emergency procedure 2.2 (1.0 – 4.5) 0.768 8

High or intermediate risk for postoperative pulmonary complications ≥ 26

CI: confidence interval; OR: odds ratio; SpO2: oxyhemoglobin saturation by pulse oximetry breathing air in supine position; g/dL: gram per decilitre†The simplified risk score was the sum of each logistic regression coefficient multiplied by 10, after rounding off its value

PROVHILO trial

Chap

ter

8

201

Table S3. Mechanical ventilation protocol for higher and lower PEEP groups

Mechanical ventilation settings Value

Ventilator Mode Volume controlled; just before tracheal extubation pressure support was allowed

Tidal Volume 8 ml/kg of predicted body weight

Inspiration : Expiration time 1:2

Respiratory Rate goal adjusted to achieve FE’CO2 between 35–45 mmHg

Fractional Inspired Oxygen 0.40 or higher to reach target SpO2 ≥ 92%

Rescue strategy with higher PEEP group

Step 1 2 3 4 5 6 7 8

FIO2 0.5 0.5 0.5 0.5 0.6 0.7 0.8 0.8

PEEP 12 10 8 6 6 6 6 4 or lower

Rescue strategy with lower PEEP group

Step 1 2 3 4 5 6 7 8 9

FIO2 0.5 0.6 0.6 0.6 0.6 0.7 0.8 0.8 RM

PEEP 2 2 3 4 5 5 5 6 6

Recruitment manoeuvre in higher PEEP group

Set peak inspiratory pressure limit at 45 cmH2OKeep tidal volume at 8 ml/kg PBW, PEEP at 12 cmH2O and inspiration : expiration time unchangedSet RR to 6–8 breaths/min, or lowest RR that ventilator allows Increase tidal volume in steps of 4 ml/kg of PBW, continue until plateau pressure = 30–35 cmH2O, and hold mechanical ventilation settings for 3 breathsSet RR and tidal volume back to values preceding the recruitment maneuver, keep PEEP at 12 cmH2O

FE’CO2: end–tidal carbon dioxide partial pressure; mmHg: millimetre of mercury; SpO2: oxyhaemoglobin saturation measured by pulse oximeter; FIO2: fraction inspired oxygen; RM: recruitment manoeuvre; PBW: predicted body weight, calculated according to a predefined formula: 50 + 0.91 x (centimetres of height – 152.4) for males and 45.5 + 0.91 x (centimetres of height – 152.4) for females; PEEP: positive end-expiratory pressure; RR: Respiratory Rate

202

Table S4. Definitions of pulmonary postoperative complications

Postoperative pulmonary complications

Hypoxemia

PaO2 < 60 mmHg or SpO2 < 90% in room air, but responding to supplemental oxygen (excluding hypoventilation)

Severe hypoxemia

Need for non–invasive or invasive mechanical ventilation or a PaO2 < 60 mmHg or SpO2 < 90% despite supplemental oxygen (excluding hypoventilation)

Bronchospasm

Defined as newly detected expiratory wheezing treated with bronchodilators

Suspected pulmonary infection

In case patient receives antibiotics and meets at least one of the following criteria: new or changed sputum, new or changed lung opacities on chest X–ray when clinically indicated, tympanic temperature > 38.3°C, WBC count > 12 x109/L

Pulmonary infiltrate

Chest X–ray demonstrating monolateral or bilateral infiltrate

Aspiration pneumonitis

Defined as respiratory failure after the inhalation of regurgitated gastric contents

Acute Respiratory Distress Syndrome

By the consensus criteria (only in case of non–invasive or invasive mechanical ventilation)2

Atelectasis

Suggested by lung opacification with shift of the mediastinum, hilum, or hemidiaphragm towards the affected area, and compensatory overinflation in the adjacent nonatelectatic lung

Pleural effusion

Chest X–ray demonstrating blunting of the costophrenic angle, loss of the sharp silhouette of the ipsilateral hemidiaphragm in upright position, evidence of displacement of adjacent anatomical structures, or (in supine position) a hazy opacity in one hemi–thorax with preserved vascular shadows

Pulmonary oedema caused by cardiac failure

Defined as clinical signs of congestion, including dyspnoea, oedema, rales and jugular venous distention, with the chest X–ray demonstrating increase in vascular markings and diffuse alveolar interstitial infiltrates

Pneumothorax

Defined as air in the pleural space with no vascular bed surrounding the visceral pleura

PROVHILO trial

Chap

ter

8

203

Table S5. Definitions of extra-pulmonary postoperative complications

Extra-pulmonary postoperative complications

Systemic inflammatory response syndrome (SIRS)3

Presence of two or more of the following findings: body temperature < 360C or > 380C; heart rate > 90 beats per minute; respiratory rate > 20 breaths per minute or on blood gas PaCO2 < 32 mmHg (4.3 kPa); WBC count < 4 x109/L or >12 x109/L, or > 10% band forms

Sepsis

SIRS in response to a confirmed infectious process; infection can be suspected or proven (by culture, stain, or polymerase chain reaction (PCR)), or a clinical syndrome pathognomonic for infection. Specific evidence for infection includes WBCs in normally sterile fluid (such as urine or cerebrospinal fluid), evidence of a perforated viscus (free air on abdominal X–ray or CT scan, signs of acute peritonitis), abnormal chest X–ray consistent with pneumonia (with focal opacification), or petechiae, purpura, or purpura fulminans

Severe sepsis

Sepsis with organ dysfunction, hypoperfusion, or hypotension

Septic shock

Sepsis with refractory arterial hypotension or hypoperfusion abnormalities in spite of adequate fluid resuscitation; signs of systemic hypoperfusion may be either end-organ dysfunction or serum lactate greater than 4 mmol/L. Other signs include oliguria and altered mental status. Patients are defined as having septic shock if they have sepsis plus hypotension after aggressive fluid resuscitation, typically upwards of 6 litres or 40 ml/kg of crystalloid

Extrapulmonary infection

Wound infection or any other infection

Coma

Glasgow Coma Score < 8 in the absence of therapeutic coma or sedation

Acute myocardial infarction4

Detection of rise and/or fall of cardiac markers (preferably troponin) with at least one value above the 99th percentile of the upper reference limit, together with: symptoms of ischemia, ECG changes indicative of new ischemia, development of pathological Q-waves, or imaging evidence of new loss of viable myocardium or new regional wall motion abnormality, or: sudden unexpected cardiac death, involving cardiac arrest with symptoms suggestive of cardiac ischemia (but death occurring before the appearance of cardiac markers in blood)

Acute renal failure (ARF)5

Renal failure documented as follows: Risk: increased creatinine x1.5 or glomerular filtration rate (GFR) decrease > 25% or urine output (UO) < 0.5 ml/kg/h x 6 h; Injury: increased creatinine x2 or GFR decrease > 50% or UO < 0.5 ml/kg/h x 12 hr; Failure: increase creatinine x3 or GFR decrease > 75% or UO < 0.3 ml/kg/h x 24 hr or anuria x 12 hrs; Loss: persistent ARF = complete loss of kidney function > 4 weeks

204

Disseminated intravascular coagulation (DIC)6

DIC score documented as follows: Platelet count < 50 (2 points), < 100 (1 point), or ≥ 100 (0 points); D–dimer > 4 µg/ml (2 points), > 0.39 µg/ml (1 point) or ≤ 0.39 µg/ml (0 points); Prothrombin time > 20.5 seconds (2 points), > 17.5 seconds (1 point) or ≤ 17.5 seconds (0 points); if ≥ 5 points: overt DIC

Hepatic failure

Serum bilirubin level > 34 µmol/L with elevation of the transaminase and lactic dehydrogenase levels above twice normal values

Gastro–intestinal failure7

Gastro–intestinal bleeding

Gastro–intestinal failure (GIF) score documented as follows: 0 = normal gastrointestinal function; 1 = enteral feeding with under 50% of calculated needs or no feeding 3 days after abdominal surgery; 2 = food intolerance (FI) or intra–abdominal hypertension (IAH); 3 = FI and IAH; and 4 = abdominal compartment syndrome (ACS)

Impaired wound healing

Interruption in the timely and predictable recovery of mechanical integrity in the injured tissue

Table S6. Specification recruitment manoeuvres in the higher PEEP group

Condition % (n/N) Reasons

All RM as indicated by the protocol 86 (378/442)

Neither RMs after intubation nor before extubation 1 (6/442)

Misunderstanding of the study protocol by the local investigator (n = 3), RM after intubation not performed because of hypotension, RM before extubation not performed because of massive bleeding (n = 1); reason not recorded (n = 2)

No RM before extubation, but RM after intubation performed 13 (58/442)

Forgotten (n = 3), logistic reasons (n = 2), misunderstanding of the study protocol by the local investigator (n = 17), reason not recorded (n = 36)

RM: recruitment manoeuvre

PROVHILO trial

Chap

ter

8

205

Table S7a. Specification of rescue strategy for desaturation

Total subjects Maintained PEEP/FiO2

modificationShortPEEP/FiO2 modification

Lower PEEP 34 23 11

Higher PEEP 11 7 4

Table S7b. Highest step of rescue strategy for desaturation

Step n

Lower PEEP 1 5

N = 34 2 8

3 1

4 0

5 9

6 4

7 0

8 1

9 0

Not recorded 6

Higher PEEP 1 4

N = 11 2 2

3 1

4 1

5 1

6 0

7 0

8 0

Not recorded 2

The duration modification of FiO2 and/or PEEP was left to the discretion of to the attending anaesthesiologist.

In the lower PEEP group 11 out of 34 patients received short FiO2 and/or PEEP modifications, after which the ventilator settings were resumed according to the protocol:

- 5 patients received modification during less than 1 hour - 3 patients received modification during one hour - 3 patients received modification during two hours

In the higher PEEP group 3 out of 11 patients received short FiO2 and/or PEEP modifications: - 3 patients received modification during less than 1 hour - 1 patient received modification during one hour

206

Table S8. Postoperative surgical complications*

Variable – % (n/N) Higher PEEPn=445

Lower PEEPn=449 p value

Bleeding surgical site 16 (71/444) 13 (58/58446) 0.21

Anastomosis bleeding 2 (8 /400) 1 (5/449) 0.29

Anastomosis leakage 0.7 (3/429) 0.4 (2/449) 0.62

Anastomosis necrosis 4 (18/445) 4 (20/449) 0.76

Fistulation 0.2 (1/445) 0.2 (1/449) 1.0

Other 2 (11/445) 1.7 (8/449) 0.47

*Reported surgical complications on postoperative day one to day of discharge

Table S9. Intraoperative medication

Higher PEEPn=445

Lower PEEPn=449 p value

Anaesthetic drug – % (n/N)

Desflurane inhalation 18 (74/403) 20 (83/406) 0.45

Sevoflurane inhalation 79 (318/403) 78 (318/406) 0.95

Isoflurane inhalation 8 (33/403) 7 (27/406) 0.40

Neuromuscular blocking agent – % (n/N)

Atracurium 29 (116/403) 29 (126/428) 0.84

Cis-atracurium 39 (159/403) 37 (160/428) 0.54

Rocuronium 34 (135/403) 34 (144/428) 0.96

Intravenous opioid –% (n/N)

Fentanil 48 (208/437) 50 (219/437) 0.46

Sufentanil 43 (186/437) 43 (186/437) 1

Remifentanil 35 (154/437) 33 (145/437) 0.52

PROVHILO trial

Chap

ter

8

207

Table S10a. Per protocol analysis of patients receiving complete higher PEEP strategy

Higher PEEPN=445

Lower PEEPN=449 Total

Received complete higher PEEP strategy 368 2* 370

All other patients 77$ 447 524

Two patients in the lower PEEP group were ventilated with the complete higher PEEP strategy

$Protocol deviation from higher PEEP strategy was defined as ventilation with PEEP <10 cmH2O during 2 or more subsequent time points and/or missing 1 or more recruitment manoeuvres

Table S10b. Per protocol analysis of postoperative pulmonary complications

Higher PEEP strategy% (n/N)

All other patients% (n/N) p- value RR (95% CI)

Overall 41.4% (370/894) 58.6% (524/894)

Postoperative pulmonary complications

40% (147/366) 39% (199/514) 0.66 1.04 (0.88 – 1.22)

208

Table S11. Univariate analysis of factors associated with the primary outcome

Characteristics PPC = 0n = 534

PPC = 1n=346

Odds Ratio(95% CI) p value

Randomization: PEEP (263/534) (174/346) 0.96 (0.73 /1.26) 0.76

Age - year 64 [53-72] 68 [59 - 74] 1.02 (1.01/1.03) <0.0001

ARISCAT score 21 (114/531) 28 (97/344) 1.44 (1.05/1.96) 0.0234

Diabetes Mellitus 12 (66/534) 19 (65/346) 1.64 (1.13/2.38) 0.0093

Duration of surgery, minutes 175 [130 - 240] 250 [180 - 335] 1 (1.005/1.008) <0.0001

Respiratory rate, breaths/min 11 (1.9) 12 (2) 1.16 (1.08/1.24) < 0.0001

Minute ventilation, ml/min 5484 (1136) 5822 (1319) 1 (1/1)* <0.0001

FiO2

≤ 40% (reference) 10 (54/533) 8 (29/344) 1.22 (0.76/1.97) 0.40

40 - 60% 86 (459/533) 86 (297/344) 1.20 (0.76/1.96) 0.44

60 - 80% 3.6 (19/533) 4.7 (16/344) 1.57 (0.69/3.51) 0.27

≥ 80% 0.2 (1/533) 0.6 (2/344) 3.72 (0.34/82) 0.29

SpO2, 99 [98 - 100] 99 [98 -100] 1.02 (0.94/1.12) 0.61

etCO2 35 (3.5) 35 (3.6) 0.98 (0.95/1.02) 0.37

HR - bpm 70 (11.4) 70 (11.8) 1 (0.99/1.01) 0.65

Total fluids

< 1000 mL 6.5 (34/524) 1.8 (6/338) 3.83 (1.59/9.24) 0.0013

1000 - 3000 mL 56 (294/524) 40 (135/338) 2.60 (1.14/7) 0.0355

3000 - 5000 30 (157/524) 39 (131/338) 4.72 (2.06/12) 0.0007

> 5000 mL 7.4 (39/524) 20 (66/338) 9.59 (3.92/27) <0.0001

Platelet transfused, no. 0 (0/534) 3.8 (13/345) NA NA

Blood loss 300 [200 - 600] 600 [300 - 1200] 1 (1/1)** <0.0001

Data is presented as means ± (sd), median [IQR] or proportion % (n/N); n: number of patients; N: total patients and relative risk with 95% Confidence Intervals; PPCs: postoperative pulmonary complications; PPC = 0: no development of PPCs; PPC = 1: development of 1 or more PPCs.Variables tested in the model were selected if the p value was <0.20 and were clinically relevant parameters * actual values are OR 1.0002278 (1.00011586/1.000342) Odds Ratio for one change in unit (ml) minute ventilation **actual values are OR 1.0008824 (1.000546/1.0012656) Odds Ratio for one change in unit (ml) bloodloss

PROVHILO trial

Chap

ter

8

209

CONSORT 2010 checklist of information to include when reporting a randomised trial

Section/Topic Item No Checklist item Reported on page

No

Title and abstract

1a Identification as a randomized trial in the title 235

1b Structured summary of trial design, methods, results, and conclusions (for specific guidance see CONSORT for abstracts) 236

Introduction

Background and objectives

2a Scientific background and explanation of rationale 237

2b Specific objectives or hypotheses 237

Methods

Trial design3a Description of trial design (such as parallel, factorial) including

allocation ratio 238

3b Important changes to methods after trial commencement (such as eligibility criteria), with reasons n.a.

Participants4a Eligibility criteria for participants 238, 239 &

Appendix p.268, 269

4b Settings and locations where the data were collected 238 & Appendix p.263, 264

Interventions 5The interventions for each group with sufficient details to allow replication, including how and when they were actually administered

239, 240 & Appendix p.270

Outcomes6a

Completely defined pre-specified primary and secondary outcome measures, including how and when they were assessed

240 & Appendix p. 271 – 273

6b Any changes to trial outcomes after the trial commenced, with reasons n.a.

Sample size7a How sample size was determined 240, 241

7b When applicable, explanation of any interim analyses and stopping guidelines 241

Randomisation

Sequencegeneration

8a Method used to generate the random allocation sequence 239

8b Type of randomisation; details of any restriction (such as blocking and block size) 239

Allocationconcealmentmechanism

9

Mechanism used to implement the random allocation sequence (such as sequentially numbered containers), describing any steps taken to conceal the sequence until interventions were assigned

239

Implementation 10 Who generated the random allocation sequence, who enrolled participants, and who assigned participants to interventions 239

210

Section/Topic Item No Checklist item Reported on page

No

Blinding11a

If done, who was blinded after assignment to interventions (for example, participants, care providers, those assessing outcomes) and how

239

11b If relevant, description of the similarity of interventions n.a.

Statistical methods

12a Statistical methods used to compare groups for primary and secondary outcomes 241

12b Methods for additional analyses, such as subgroup analyses and adjusted analyses 241

Results

Participant flow (a diagram is strongly recommended)

13aFor each group, the numbers of participants who were randomly assigned, received intended treatment, and were analysed for the primary outcome

242, 256, 257

13b For each group, losses and exclusions after randomisation, together with reasons 242, 256, 257

Recruitment14a Dates defining the periods of recruitment and follow-up 242

14b Why the trial ended or was stopped n.a.

Baseline data 15 A table showing baseline demographic and clinical characteristics for each group 249 - 251

Numbers analysed 16

For each group, number of participants (denominator) included in each analysis and whether the analysis was by original assigned groups

242, 249-258

Outcomes and estimation

17aFor each primary and secondary outcome, results for each group, and the estimated effect size and its precision (such as 95% confidence interval)

242, 254, 255

17b For binary outcomes, presentation of both absolute and relative effect sizes is recommended n.a.

Ancillary analyses 18Results of any other analyses performed, including subgroup analyses and adjusted analyses, distinguishing pre-specified from exploratory

238, 242 – 246, 276 & Appendix p. 274 – 279

Harms 19 All important harms or unintended effects in each group (for specific guidance see CONSORT for harms)

242, 243, 254,255 & Appendix p.275, 276

Discussion

Limitations 20 Trial limitations, addressing sources of potential bias, imprecision, and, if relevant, multiplicity of analyses 245, 246

Generalizability 21 Generalizability (external validity, applicability) of the trial findings 244 – 246

Interpretation 22 Interpretation consistent with results, balancing benefits and harms, and considering other relevant evidence 245, 246

PROVHILO trial

Chap

ter

8

211

Other information

Registration 23 Registration number and name of trial registry 236, 238

Protocol 24 Where the full trial protocol can be accessed, if available 238

Funding 25 Sources of funding and other support (such as supply of drugs), role of funders 236, 242

CONSORT Checklist for Reporting Randomized Controlled Trials in Journal and Conference Abstracts*

Item Description Reported on line number

Title Identification of the study as randomized p.235

Authors * Contact details for the corresponding author p.235

Trial design Description of the trial design (e.g. parallel, cluster, non-inferiority) p.236, line 9, 10

Methods

Participants Eligibility criteria for participants and the settings where the data were collected p. 236, line 9 - 13

Interventions Interventions intended for each group p. 236, line 11 - 12

Objective Specific objective or hypothesis p. 236, line 6 – 8

Outcome Clearly defined primary outcome for this report p. 236, line 14 - 15

Randomization How participants were allocated to interventions p. 236, line 13

Blinding (masking)Whether or not participants, care givers, and those assessing the outcomes were blinded to group assignment

p. 236, line 14

Results

Numbers randomized Number of participants randomized to each group p. 236, line 17 - 18

Recruitment Trial status n.a.

Numbers analysed Number of participants analysed in each group p. 236, line 21 - 22

Outcome For the primary outcome, a result for each group and the estimated effect size and its precision p. 236, line 21 - 22

Harms Important adverse events or side effects p. 236, line 23 - 24

Conclusions General interpretation of the results p. 236, line 1 – 2

Trial registration Registration number and name of trial register p. 236, line 15, 16

Funding Source of funding p. 236, line 26, 27

*This item is specific to conference abstracts

212

References1. Canet J, Gallart L, Gomar C, et al. Prediction of Postoperative Pulmonary Complications in a Population-based Surgical

Cohort. Anesthesiology 2010; 113: 1338-502. Bernard GR, Artigas A, Brigham KL, et al. Report of the American-European consensus conference on ARDS: definitions,

mechanisms, relevant outcomes and clinical trial coordination. The Consensus Committee. Intensive Care Med 1994; 20: 225-32

3. Bone RC. Toward an epidemiology and natural history of SIRS (systemic inflammatory response syndrome). JAMA 1992; 268: 3452-5

4. Thygesen K, Alpert JS, White HD, et al. Universal definition of myocardial infarction. Circulation 2007; 116: 2634-535. Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky P. Acute renal failure – definition, outcome measures, animal

models, fluid therapy and information technology needs: the Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care 2004; 8: R204-12

6. Levi M, Toh CH, Thachil J, Watson HG. Guidelines for the diagnosis and management of disseminated intravascular coagulation. British Committee for Standards in Haematology. Br J Haematol 2009; 145: 24-33

7. Reintam A, Parm P, Kitus R, Starkopf J, Kern H. Gastrointestinal failure score in critically ill patients: a prospective observational study. Crit Care 2008; 12: R90

PROVHILO trial

Chap

ter

8

213

Chapter 9

Positive end-expiratory pressure during surgery - Authors’ reply

Hemmes SNT, Gama de Abreu M, Pelosi P, Schultz MJLancet 2014; 8; 384(9955):1670-1

216

Positive end-expiratory pressure during surgery – Comments Hans-Joachim Priebe (Department of Anaesthesia, University Hospital Freiburg, Germany)

With great interest, I read the Article by PROVE Network Investigators (Aug 9, p 495)1 and the accompanying Comment.2 Some of the study limitations that might have obscured any treatment effect (e.g., protocol deviations, infrequent recruitment manoeuvres, and a high positive end-expiratory pressure [PEEP] level in the higher PEEP group) have been addressed in the accompanying Comment.2 Several additional factors might have negated any potential treatment effect.

39% of patients in both groups had received thoracic epidural analgesia. Compared with systemic analgesia, thoracic epidural analgesia can be expected to improve the postoperative pulmonary outcome in patients undergoing abdominal surgery.3 At the same time, the combination of thoracic epidural analgesia, general anesthesia, and mechanical ventilation frequently causes hypotension, which requires therapy.4 Any possible difference in pulmonary outcome between groups might have been obscured by the beneficial pulmonary effects of thoracic epidural analgesia. Similarly, the higher incidence of intraoperative hypotension and increased need for vasoactive drugs in the high compared with the low PEEP group might not have mainly been caused by the high PEEP per se but instead by the combination of high PEEP, thoracic epidural analgesia, and general anaesthesia. To compare the primary and secondary outcome variables between patients with and without thoracic epidural analgesia would be relevant. Combined abrupt withdrawal of 12 cmH2O PEEP and restoration of spontaneous respiration at the time of extubation will have acutely increased venous return and, in turn, right and left ventricular preload. This might have increased lung water in patients with left ventricular dysfunction with unpredictable subsequent adverse pulmonary sequelae.

All patients had received intermediate long acting muscle relaxants. Residual neuromuscular blockade must be expected at the end of surgery in up to 80% of cases.5 Residual neuromuscular blockade is associated with impaired postoperative lung function and postoperative pulmonary morbidity.5,6 Because the detrimental effect of residual neuromuscular blockade on postoperative pulmonary outcome might have obscured any potential treatment effects, we need to know whether neuromuscular function was quantitatively assessed before extubation. Extubation during an inspired oxygen fraction (FiO2) of 1.0 is associated with worse postextubation atelectasis and oxygenation compared with extubation at a lower FiO2.7 Use of a FiO2 of 1.0 at the time of extubation in all patients might partly explain the absence of difference in postoperative atelectasis.

PROVHILO Authors’ reply

Chap

ter

9

217

References1. The PROVE Network Investigators for the Clinical Trial Network of the European Society of Anaesthesiology. High

versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial. Lancet 2014; 384: 495–503

2. Futier E. Positive end-expiratory pressure in surgery: good or bad? Lancet 2014; 384: 472–743. Popping DM, Elia N, Marret E, Remy C, Tramèr MR. Protective effects of epidural analgesia on pulmonary complications

after abdominal and thoracic surgery. A metaanalysis. Arch Surg 2008; 143: 990–994. De Kock M, Laterre P-F, Andruetto P, et al. Ornipressin: an efficient alternative to counteract hypotension during

combined general/epidural anesthesia. Anesth Analg 2000; 90: 1301–075. Plaud B, Debaene B, Donati F, Marty J. Residual paralysis after emergence from anesthesia. Anesthesiology 2010; 112:

1013–226. Berg H, Viby-Mogensen J, Roed J, et al. Residual neuromuscular block is a risk factor for postoperative pulmonary

complications: A prospective, randomised, and blinded study of postoperative pulmonary complications after atracurium, vecuronium and pancuronium. Acta Anaesthesiol Scand 1997; 41: 1095–103

7. Benoit Z, Wicky S, Fischer J-F, et al. The effect of increased FiO2 before tracheal extubation on postoperative atelectasis. Anesth Analg 2002; 95: 1777–81

218

Positive end-expiratory pressure during surgery – Comments Farouk Mike Elkhatib, Mohamad Khatib (Department of Anesthesiology, School of Medicine (FME), American University of Beirut, Beirut, Lebanon)

We read with interest the PROVHILO study1 comparing high with low positive end-expiratory pressure (PEEP) during general anaesthesia for open abdominal surgery. The researchers concluded that high PEEP and recruitment manoeuvres during open abdominal surgery do not protect against postoperative pulmonary complications.1 We commend the investigators for providing such highly needed data; however, we believe that a major contributing factor that could have masked the potential benefit of high PEEP is the fact that peak inspiratory pressure was significantly higher in patients exposed to high PEEP than those exposed to low PEEP. The fact that high-peak airway and alveolar pressures per se are associated with ventilator-induced lung injury secondary to alveolar overdistention is well known.2

The PROVHILO study1 should have considered decreasing the tidal volume (VT) in the high-PEEP group to whatever level is needed to achieve in the high-PEEP group a peak airway pressure that is similar to that of the low-PEEP group. As such, one would expect higher PEEP with lower VT to maintain the same peak airway pressure that might result in improved lung compliance secondary to enhanced alveolar recruitment with high PEEP while preventing hyperinflation or overdistention in non-dependent lung units by the decrease in VT.3 Furthermore, reduction in peak airway pressure in the high PEEP group will lead to a decrease in mean airway pressure and potentially less haemodynamic impairment in the high-PEEP group.4 Perhaps only prevention of all forms of ventilator-induced lung injury (ie, cyclic opening and closing of alveoli and alveolar overdistention) can reduce or eliminate complications in mechanically ventilated patients.5

References1. The PROVE Network Investigators for the Clinical Trial Network of the European Society of Anaesthesiology. High

versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial. Lancet 2014; 384: 495–503

2. Futier E, Constantin JM, Paugam-Burtz C, et al, and the IMPROVE Study Group. A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. NEJM 2013;369: 428–37

3. Severgnini P, Selmo G, Lanza C, et al. Protective mechanical ventilation during general anesthesia for open abdominal surgery improves postoperative pulmonary function. Anesthesiology 2013; 118: 1307–21

4. Luecke T, Pelosi P. Clinical review: Positive end-expiratory pressure and cardiac output. Crit Care 2005; 9: 607–215. Melo MF, Eikermann M. Protect the lungs during abdominal surgery: it may change the postoperative outcome.

Anesthesiology 2013; 118: 1254–57

PROVHILO Authors’ reply

Chap

ter

9

219

Authors’ reply Emmanuel Futier,1 Farouk Mike Elkathib and Mohamad Kathib, and Hans-Joachim Priebe’s comments about the results of PROVHILO2 raise relevant issues that deserve consideration.

Firstly, PROVHILO is the only trial of intraoperative ventilation that compared different levels of positive end-expiratory level (PEEP) in low tidal volume ventilation, which is now the standard of care.3,4 Notably, results from one metaanalysis5 show that the benefit from lung-protective ventilation is better explained by the use of low tidal volumes than the use of high levels of PEEP. A simultaneous reduction in tidal volume size in the high PEEP group in PROVHILO, to limit peak airway pressures, would not only have contradicted current practice but also have made identification of the individual role of high levels of PEEP impossible. Although we agree that high airway pressures could lead to increased ventilator-induced lung injury, mean peak inspiratory pressures were 23 cm H2O in the high PEEP group, i.e. below the commonly accepted safety limit.

Differently from the claim by Futier,1 several trials show that an intraoperative PEEP level of 10 cmH2O or more is necessary to avoid lung volume loss, and is not associated with overdistension.6 In PROVHILO, dynamic compliance improved in the high PEEP group, suggesting effective lung recruitment without relevant overdistension. Furthermore, periodic recruitment manoeuvres7 are not considered essential to keep lungs open when appropriate levels of PEEP are used, even with use of high inspiratory oxygen concentrations.8 Also, the suggestion that high oxygen concentrations during extubation could induce postoperative atelectasis has recently been challenged.9 We also do not ascribe pulmonary complications to abrupt changes in intrathoracic pressures during extubation, as a stepwise and gentle approach was used in which the mean airway pressure was reduced first, before resuming spontaneous breathing—a strategy that lasted several minutes, as usual in clinical practice.

Several additional analyses were requested. In an analysis of patients who received high levels of PEEP and received recruitment manoeuvres after intubation, after disconnection of the ventilator and before extubation (363 patients), versus patients who received low levels of PEEP without recruitment manoeuvres at any time (441 patients), the occurrence of postoperative pulmonary complications was not different (35% vs 36%; p=0.80). Likewise, in the 445 patients without thoracic epidural anaesthesia, the incidence of postoperative pulmonary complications did not differ between PEEP groups (48 vs 42%; p=0.26). Finally, in 265 patients who were monitored for and showed no residual neuromuscular blockade, the occurrence of postoperative pulmonary complications was independent of the level of PEEP (50% vs 45%; p=0.13).

Differently from a recent retrospective, one-centre study,4 results from PROVHILO1 convincingly showed that in non-obese patients undergoing open abdominal surgery, a low tidal volume strategy combined with low levels of PEEP is not associated with any disadvantage in terms of clinical outcome. In fact, use of high levels of PEEP was associated with impaired hemodynamics, mandating the increased use of fluids and vasoactive drugs in the intraoperative period.

PROVHILO clearly answers Futier’s question “positive end-expiratory pressure in surgery: good or bad?” in the patient group being studied. When a low tidal volume strategy is used, a low

220

PEEP level without recruitment manoeuvres does not increase the incidence of postoperative pulmonary complications and is associated with less haemodynamic impairment compared with high PEEP with recruitment manoeuvres. These results need to be confirmed in morbidly obese patients and in patients undergoing thoracic surgery. We hope that beliefs and myths regarding PEEP during general anaesthesia will soon give place to the evidence of large randomised controlled trials.

References1. Futier E, Constantin JM, Paugam-Burtz C, et al, and the IMPROVE Study Group. A trial of intraoperative low-tidal-volume

ventilation in abdominal surgery. NEJM 2013; 369: 428–372. The PROVE Network Investigators for the Clinical Trial Network of the European Society of Anaesthesiology. High

versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial. Lancet 2014; 384: 495–503

3. Jaber S, Coisel Y, Chanques G, et al. A multicentre observational study of intra-operative ventilatory management during general anaesthesia: tidal volumes and relation to body weight. Anaesthesia 2012; 67: 999–1008

4. Levin MA, McCormick PJ, Lin HM, Hosseinian L, Fischer GW. Low intraoperative tidal volume ventilation with minimal PEEP is associated with increased mortality. Br J Anaesth 2014; 113: 97–108

5. Serpa Neto A, Hemmes SN, Barbas CS, Gama de Abreu M, Pelosi P, Schultz MJ. Intraoperative ventilator settings and postoperative acute respiratory distress syndrome: an individual data metaanalysis of 3 659 patients. ATS 2014 International Conference; San Diego; May 16–21, 2014

6. Satoh D, Kurosawa S, Kirino W, et al. Impact of changes of positive end-expiratory pressure on functional residual capacity at low tidal volume ventilation during general anesthesia. J Anesth 2012; 26: 664–69

7. Slutsky AS, Ranieri VM. Ventilator-induced lung injury. NEJM 2013; 369: 2126–368. Neumann P, Rothen HU, Berglund JE, Valtysson J, Magnusson A, Hedenstierna G. Positive end-expiratory pressure

prevents atelectasis during general anaesthesia even in the presence of a high inspired oxygen concentration. Acta Anaesthesiol Scand 1999; 43: 295–301

9. Edmark L, Auner U, Lindback J, Enlund M, Hedenstierna G. Post-operative atelectasis—a randomised trial investigating a ventilatory strategy and low oxygen fraction during recovery. Acta Anaesthesiol Scand 2014; 58: 681–88

PROVHILO Authors’ reply

Chap

ter

9

221

Chapter 10

Incidence of mortality and morbidity related to postoperative lung injury in patients who have undergone abdominal or thoracic surgery: a systematic review and metaanalysis

Serpa Neto A, Hemmes SNT, Barbas CS, Beiderlinden M, Fernandez-Bustamante A, Futier E, Hollmann MW, Jaber S, Kozian A, Licker M, Lin WQ, Moine P, Scavonetto F, Schilling T, Selmo G, Severgnini P, Sprung J, Treschan T, Unzueta C, Weingarten TN, Wolthuis EK, Wrigge H, Gama de Abreu M, Pelosi P, Schultz MJ for the PROVE Network investigators. Lancet Respiratory Medicine 2014; 2(12):1007-15

224

Abstract

Background. Lung injury is one of the most serious complications following surgery. Development of postoperative lung injury and its effect on outcome could depend on ventilator settings during the surgical procedure. We performed a systematic review and metaanalysis to estimate the incidence and determined the crude and attributable morbidity and in-hospital mortality of postoperative lung injury in published investigations of intra-operative ventilation for abdominal or thoracic surgery.

Methods. Observational studies and randomized controlled trials were identified by a systematic search of MEDLINE, CINAHL, Web of Science, and CENTRAL and screened for inclusion into a metaanalysis. Key inclusion criteria were: adults; ventilation for general anesthesia for abdominal or thoracic surgery; use of protective versus conventional ventilation. Individual patient data were obtained from the corresponding authors. Attributable mortality was calculated subtracting the in-hospital mortality rate of patients who did not develop postoperative lung injury from those who developed postoperative lung injury in predefined patient groups.

Findings. Data from 12 investigations were included, comprising 3,365 patients. The total incidence of postoperative lung injury in abdominal and thoracic surgery patients was similar (3.4 vs. 4.3%; p = 0.198). Patients who developed postoperative lung injury were older, had higher ASA scores, higher prevalence of sepsis or pneumonia, more frequently had received blood transfusions during surgery, and ventilation with higher tidal volumes and/or lower positive end-expiratory pressure levels. The occurrence of postoperative lung injury was associated with longer stay in intensive care unit and hospital (8 ± 13 vs. 1 ± 4 days; p < 0.001, and 21 ± 18 vs. 15 ± 14 days; p < 0.001, respectively), and increased in-hospital mortality (20.3 vs. 1.4%; p < 0.001). Overall attributable mortality of postoperative lung injury was 19% [95% confidence interval 18–19%), and differed between abdominal and thoracic surgery patients (12% vs. 27%, respectively; p < 0.001) but it was independent from intra-operative ventilator settings.

Interpretation. Development of postoperative lung injury was associated with increased in–hospital mortality and by a longer ICU and hospital lengths of stay. Attributable mortality of postoperative lung injury is higher in thoracic than in abdominal surgery patients. Protective mechanical ventilation reduces the incidence of postoperative lung injury but seems to have no effect on attributable mortality of this condition.

IPD metaanalysis postoperative lung injury

Chap

ter

10

225

Introduction

More than 230 million major surgical procedures are undertaken worldwide each year.1 Postoperative complications after major surgery increase resource use and are an important cause of death.1 Especially postoperative pulmonary complications, including postoperative lung injury, are suggested to have a strong impact on morbidity and mortality of patients who need major surgery.2-4 There is growing evidence that so–called intra–operative lung–protective mechanical ventilation with low tidal volumes and with or without high levels of positive end–expiratory pressure (PEEP) prevents postoperative lung injury compared to conventional ventilation with the use of high tidal volume levels and low levels of PEEP.2-4 Recently, in a report of a large retrospective study that showed use of low tidal volumes during general anesthesia for surgery to be associated with increased mortality, it was suggested that excess mortality could have been caused by the use of too low levels of PEEP.5

The exact impact of postoperative lung injury on morbidity and mortality is uncertain, and the outcome of postoperative lung injury could be different in patients who had abdominal surgery from those who underwent thoracic surgery. In addition, the impact of lung–protective ventilation on outcome of patients developing postoperative lung injury needs to be better defined.2-4 Indeed, it seems possible that outcome of lung injury is different according to the strategy of intraoperative ventilation.

A better understanding of the incidence, morbidity and mortality of postoperative lung injury may help design future trials, and possibly improves the approach to this condition, therefore, we performed an individual patient data metaanalysis of studies and trials of intra–operative ventilation for abdominal and thoracic surgery, which offered the possibility to quantify crude and attributable mortality of postoperative lung injury and its relationship with the strategy of ventilation used during surgery. We compared patients after abdominal surgery with patients after thoracic surgery, and related the outcome to intra–operative ventilator settings. We hypothesized that crude and attributable mortality are different between abdominal and thoracic surgery. In addition we hypothesized outcome of postoperative lung injury to be dependent on intraoperative ventilation settings.

Methods

The full statistical analysis plan of this collaborative metaanalysis has been published before.6

Search strategyTwo authors performed a computerized blinded search of MEDLINE, Cumulative Index to Nursing and Allied Health Literature (CINAHL), Web of Science, and Cochrane Central Register of Controlled Trials (CENTRAL) up to April 2014. The sensitive search strategy combined the following Medical Subject Headings and Keywords ([protective ventilation OR lower tidal volume OR low tidal volume OR positive end–expiratory pressure OR positive end–expiratory pressure OR PEEP]). All reviewed articles and cross–referenced studies from retrieved articles were screened for pertinent information. We ran a computerized search of proceedings of annual meetings

226

from critical care and anaesthesiology societies to identify relevant studies published in abstract form only.

Selection of studiesObservational studies and randomized controlled trials comparing different tidal volume and PEEP settings during intraoperative ventilation for surgical general anesthesia identified by the search criteria and reporting outcomes of interest were screened for inclusion with no restrictions on language. Key inclusion criteria were: adults (i.e., age > 18 years); patients receiving ventilation for general anesthesia for abdominal or thoracic surgery; use of protective versus conventional ventilation. Studies or trials were excluded from this metaanalysis if they: included patients with pre–existing lung injury; or reported on patients receiving ventilation in a non–surgical setting (i.e., continued ventilation in the intensive care unit). The Jadad score was used to assess the quality of the randomized controlled trials and the GRACE checklist to observational studies.

Collection of individual patient dataCorresponding authors of the identified eligible published studies were contacted via email with a letter detailing background and objectives of this metaanalysis, and a datasheet for input of individual patient data. The filled out data templates were sent back to the principal investigator and further communication was by email. Corresponding authors were also contacted on unpublished data, if present to enlarge the clinical data pool. The same two investigators who performed the electronic database search handled the individual patient data provided by the corresponding authors. Data was accepted in any kind of electronic format (SPSS, STATA, Word document, Excel document, and Access document) and only the coordinators of the collaboration have direct access to the data. Both authors performed data validation, checking the received data set for data entry mistakes and inconsistency. Differences were discussed and settled in consensus.

DefinitionsFor postoperative lung injury we used the definitions for acute lung injury and ARDS from the American–European Consensus criteria group (PaO2 / FiO2 < 300),7 since all studies were conducted before the publication of the Berlin definition; follow–up was defined as the time between the day of the surgery and hospital discharge, or in–hospital death. The number of ICU and hospital–free days and alive by day 28 was defined as the mean number of days on which patients were outside the ICU or hospital and alive from day 1 to day 28. Protective ventilation was defined as ventilation using low tidal volume (defined as a tidal volume ≤ 8 ml/kg predicted body weight [PBW]) with or without high levels of PEEP (defined as PEEP ≥ 5 cmH2O) and with or without recruitment manoeuvres. Conventional ventilation was defined as ventilation using high tidal volume (> 8 ml/kg PBW) with low levels of PEEP (< 5 cmH2O) and without recruitment manoeuvres. The definition of protective and conventional ventilation was made based on several reports in the literature and according to the previously published protocol.2-4

Statistical analysisAttributable morbidity was calculated by subtracting the in–hospital mortality rate of patients without postoperative lung injury from the in–hospital mortality of patients with postoperative lung injury. The incidence rate was calculated as number of cases person-years = ([number of

IPD metaanalysis postoperative lung injury

Chap

ter

10

227

cases / person-day] x [365 person-day / 1 person-year]). A random-effect model was used to pool the overall data.

Survival curves were constructed using the Kaplan–Meier methods and compared using the log–rank test. Time–to–event was defined as time from the day of surgery to the event. We used a Cox proportional–hazards regression model to examine simultaneously effects of multiple covariates on outcome, censoring a patient’s data at the time of death, hospital discharge, or after 30 days. The initial model included age, sex, body mass index, ASA (American Society of Anesthesiology score), smoking, and predisposing conditions [defined as shock, pneumonia, blood transfusion and/or sepsis]). Variables with p < 0.2 in the univariate analysis are included in the multivariate regression. The final model was developed by dropping each variable in turn from the model and conducting a likelihood-ratio test to compare the full and the nested models (stepwise backward approach). We used a significance level of 0.05 as the cut-off to exclude a variable from the model. In all models, the categorical variables were tested for trend with the absence of postoperative lung injury as reference and the proportional–hazards assumption was assessed. A test for interaction between pairs of variables in the final model was performed. Interaction between biological plausible variable was assessed. The effect of each variable in these models was assessed with the use of the Wald test and described by the HR with 95% CI.

Subgroup analyses were performed to examine the effects of intra–operative ventilation (conventional vs. lung–protective ventilation), age (< 65 vs. ≥ 65 years), surgery (abdominal vs. thoracic) and severity of illness (ASA score < 3 or ≥ 3).

All analyses were conducted with SPSS v.20 (IBM Corporation, New York, USA) and R v.2.12.0 (R Foundation for Statistical Computing, Vienna, Austria). For all analyses two–sided p values < 0.05 were considered significant.

Results

Search results and collection of individual patient dataThe search identified three observational studies and 21 randomized controlled trials comparing different tidal volume and/or PEEP settings in intra–operative ventilation during general anaesthesia for major surgery.8-31 We were not able to collect data from five randomized controlled trials due to the following reasons: the corresponding author could not provide data of interest or had no longer access to the complete database (n = 3);20-22 or the corresponding author could not be contacted (n = 2).23,24 Seven papers were excluded because assessed patients under cardiac (n = 6)25-30 or orthopaedic surgery (n = 1).31 The total enrolment based on the studies and trials for which individual patient data could be collected was 3,365 patients (table 1, Appendix figure 1, Appendix tables 2, Appendix table 3, and Appendix table 4).

Characteristics of patients who did or did not developed postoperative lung injuryBaseline characteristics and predisposing conditions differed between patients who did and those who did not develop postoperative lung injury (table 2). Patients who developed postoperative

228

Tabl

e 1.

Cha

ract

eris

tics

of in

clud

ed st

udie

s

Stud

yYe

arCo

untr

yD

esig

n of

St

udy

Jada

d Sc

ore

Surg

ery

Tota

l Num

ber o

f Pati

ents

Post

op L

ung

Inju

ryM

orta

lity

Prot

ect

Conv

Prot

ect

Conv

Prot

ect

Conv

Wrig

ge e

t al7

2004

Ger

man

yRC

T2

Abd

/ Tho

*29

(47%

)33

(53%

)0

(0%

)3

(9%

)0

(0%

)0

(0%

)

Schi

lling

et a

l820

05G

erm

any

RCT

2Th

orac

ic*

75 (6

8%)

35 (3

2%)

0 (0

%)

0 (0

%)

0 (0

%)

0 (0

%)

Wol

thui

s et a

l920

08Du

tch

RCT

3Ab

dom

inal

24 (5

2%)

22 (4

8%)

0 (0

%)

2 (9

%)

0 (0

%)

0 (0

%)

Lin

et a

l1020

08Ch

ina

RCT

1Th

orac

ic*

50 (4

9%)

52 (5

1%)

1 (2

%)

14 (2

7%)

0 (0

%)

1 (2

%)

Lick

er e

t al11

2009

Swiss

Non

-RCT

---Th

orac

ic*

558

(51%

)53

3 (4

9%)

15 (3

%)

28 (5

%)

13 (2

%)

15 (3

%)

Wei

ngar

ten

et a

l1220

10U

SARC

T3

Abdo

min

al20

(50%

)20

(50%

)0

(0%

)1

(5%

)1

(5%

)0

(0%

)

Fern

ande

z-Bu

stam

ante

et a

l1320

11U

SAN

on-R

CT---

Abdo

min

al15

4 (3

6%)

275

(64%

)7

(4%

)14

(5%

)3

(2%

)3

1(%

)

Tres

chan

et a

l1420

12G

erm

any

RCT

5Ab

dom

inal

50 (4

9%)

51 (5

1%)

1 (2

%)

0 (0

%)

3 (6

%)

5 (1

0%)

Unz

ueta

et a

l1520

12Sp

ain

RCT

2Th

orac

ic*

40 (1

00%

)---

0 (0

%)

---1

(2%

)---

Seve

rgni

ni e

t al16

2013

Italy

RCT

5Ab

dom

inal

28 (5

1%)

27 (4

9%)

9 (3

2%)

16 (5

9%)

0 (0

%)

0 (0

%)

Futie

r et a

l1720

13Fr

ance

RCT

5Ab

dom

inal

200

(50%

)20

0 (5

0%)

1 (0

.5%

)8

(4%

)6

(3%

)7

(3%

)

Hem

mes

et a

l1820

14M

ulti*

*RC

T5

Abdo

min

al88

9 (1

00%

)---

7 (0

.8%

)---

14 (2

%)

---Co

nv.:

conv

entio

nal:

Abd:

abd

omin

al; T

ho: t

hora

cic;

RCT

: ran

dom

ized

cont

rolle

d tr

ial;

*: T

hora

cic

proc

edur

es: L

obec

tom

y (5

2%);

Pneu

mec

tom

y (2

0%);

Smal

l res

ectio

ns, l

ike

parti

al

lobe

ctom

y, a

typi

cal a

nd w

edge

rese

ction

s (20

%);

Esop

hage

ctom

y (3

%);

Oth

ers,

like

med

iasti

nal r

esec

tion

and

expl

orati

ve th

orac

otom

y (5

%);

**: E

urop

e an

d U

SA

IPD metaanalysis postoperative lung injury

Chap

ter

10

229

lung injury were older, presented higher ASA score, higher prevalence of sepsis or pneumonia, more frequently had received blood transfusion, and were ventilated with higher tidal volumes and/or lower PEEP levels (table 2). Baseline characteristics stratified according to the type of surgery are shown in the Appendix table 5 and Appendix table 6. Ventilatory parameters and duration of ventilation in patients ventilated with a protective or conventional strategy of ventilation are shown in Appendix table 7.

Incidence and timing of postoperative lung injuryThe incidence of postoperative lung injury in the whole cohort was 3.9% (crude incidence 0.99 cases per person-year). The individual and pooled postoperative lung injury incidence rates are shown in Appendix figure 8. The incidence of postoperative lung injury was higher in patients older than 65 years (4.7 vs. 2.8%; p = 0.008), and in those ventilated with conventional ventilation (6.7 vs. 2.0%; p < 0.001), while it was similar after abdominal or thoracic surgery (3.4 vs. 4.3%; p

Table 2. Patient demographics

Variable Total(n = 3365)

No PLI(n = 3150)

PLI(n = 123) p value

Age, years 62.6 ± 12.7 62.5 ± 12.7 66.4 ± 11.6 0.001

Gender, male 2019 (60.0) 1941 (61.6) 78 (63.4) 0.834

ASA 2.4 ± 0.6 2.4 ± 0.6 2.6 ± 0.6 0.003

BMI, kg/m2 25.7 ± 4.8 25.7 ± 4.9 26.1 ± 4.8 0.323

Current smoker 1107 (32.9) 1058 (33.5) 49 (39.8) 0.122

Predisposing conditions Shock Pneumonia Transfusion of blood products Sepsis

54 (1.6)25 (0.7)183 (5.4)12 (0.3)

52 (1.6)11 (0.3)168 (5.3)8 (0.2)

2 (1.6)14 (11.3)15 (12.1)4 (3.2)

0.714< 0.001< 0.001< 0.001

Ventilatory Parameters* Tidal volume, ml/kg PBW PEEP, cmH2O Respiratory rate, bpm FiO2, %

8.2 ± 1.94.4 ± 3.811.9 ± 2.744.9 ± 14.2

8.2 ± 1.84.3 ± 3.711.9 ± 2.741.4 ± 3.9

9.3 ± 2.12.9 ± 3.411.7 ± 2.240.5 ± 4.2

< 0.001< 0.0010.3840.102

Oxygenation Parameters* pH PaO2 / FiO2

PaCO2

7.3 ± 0.1320.0 ± 176.740.5 ± 6.5

7.3 ± 0.1315.0 ± 180.340.7 ± 6.7

7.4 ± 0.0302.4 ± 165.841.3 ± 3.8

0.0020.6810.708

ICU LOS, days 1.5 ± 4.8 1.1 ± 3.7 8.0 ± 12.4 < 0.001

Hospital LOS, days 15.1 ± 14.8 14.7 ± 14.3 20.9 ± 18.1 < 0.001

Mortality, % 70 (2.1) 45 (1.4) 25 (20.3) < 0.001

PLI: postoperative lung injury; BMI: body mass index; PBW: predicted body weight; BPM: breaths per minute; FiO2: inspired fraction of oxygen; ICU: intensive care unit; LOS: length of stay; PEEP: positive-end expiratory pressure*: in the middle of the surgery

230

= 0.198) (Table 3). The results stratified according to type of surgery are shown in the Appendix table 9 and Appendix table 10. The development of postoperative lung injury occurred within the first three days after surgery (mean of 2.9 ± 2.2 days; median and interquartile range of 2.0 [2.0 – 3.0] days); in 22% of cases postoperative lung injury developed after the first postoperative day (table 3 and figure 1).

Outcomes of postoperative lung injuryOutcome data for the study cohort are shown in table 3 and 4. Development of postoperative lung injury was associated with an increased in–hospital mortality (20.3 vs. 1.4% in patients with or without postoperative lung injury, respectively; p < 0.001) and by a longer ICU (8 ± 13 vs. 1 ± 4 days; p < 0.001) and hospital (21 ± 18 vs. 15 ± 14 days; p < 0.001) lengths of stay. The number of ICU–free days at day 28 and hospital–free days at day 28 were lower in patients who developed postoperative lung injury (21 ± 8 vs. 27 ± 2 days; p < 0.001; and 10 ± 7 vs. 15 ± 7 days; p < 0.001). When adjusted for age, severity of illness using ASA score, smoking, and predisposing conditions (sepsis, pneumonia, transfusion and/or shock), development of postoperative lung injury markedly increased the risk of in–hospital mortality (adjusted hazard ratio [HR], 9.58; 95% CI, 5.32 – 17.34) (figure 2 & 3, table 4 and Appendix figure 11). Patients who developed postoperative lung injury had a lower chance per day for ICU discharge, as represented by the adjusted HR of 0.45 (95% CI, 0.33 – 0.66) (table 4). The estimate of attributable mortality due to postoperative lung injury was 19% (95% CI, 18.0 – 19.1%).

In the group of patients who did develop postoperative lung injury, total duration of mechanical ventilation, ICU and hospital length of stay was similar between survivors and non–survivors

Figure 1. Timing of postoperative lung injury development during hospital stayThe day of surgery is marked as Day 0

IPD metaanalysis postoperative lung injury

Chap

ter

10

231

(Appendix table 12). The mean time to death was 35.2 ± 63.4 days and the median was 18.0 (10.0 – 31.0) days. Patients ventilated with conventional strategy during surgery died earlier than those ventilated with protective strategy (17.8 ± 13.1 vs. 51.2 ± 84.3 days; p = 0.027).

Outcomes of postoperative lung injury in predefined subgroupsThe attributable mortality of postoperative lung injury was lower in patients undergoing abdominal surgery (12.2% [95% CI, 12.0 – 12.6%]) compared to those undergoing thoracic surgery (26.5% [95% CI, 26.2 – 27.0%]) (p < 0.001). Development of postoperative lung injury was associated with a longer ICU and hospital lengths of stay, which was similar in abdominal and thoracic surgery patients (Appendix table 5 and Appendix table 6). When adjusted for age, severity of illness using ASA score, smoking, and predisposing conditions (sepsis, pneumonia, transfusion and/or shock), development of postoperative lung injury was lower in those receiving protective ventilation during surgery (adjusted HR, 0.31; 95% CI, 0.19 – 0.45). However, in–hospital mortality was independent of the ventilation strategy used in the operation room (adjusted HR, 0.71; 95% CI, 0.41 – 1.22). Attributable mortality of postoperative lung injury was similar in patients under protective ventilation (18.5% [95% CI, 17.8 – 19.2%]) compared to those under conventional ventilation (19.3% [95% CI, 19.0 – 19.7%]) (p = 0.359). Notably, patients who received conventional ventilation and developed postoperative lung injury died earlier compared to patients who received lung–protective ventilation and developed postoperative lung injury (18 ± 16 vs. 35 ± 53 days; p = 0.018). Characteristics of patients with postoperative lung injury stratified according to the type of ventilation used during surgery are shown in Appendix table

Figure 2. Kaplan–Meier estimates of the probability of overall survivalData for the Kaplan–Meier estimates of the probability of overall survival in patients without postoperative lung injury (black solid line), and patients with postoperative lung injury (black dotted line). Data were censored at 30 days after surgery

232

Tabl

e 3.

Inci

denc

e of

pos

tope

rativ

e lu

ng in

jury

and

its

char

acte

ristic

s in

all

grou

ps o

f pati

ents

*

Num

ber o

f Pati

ents

ICU

LO

S (d

ays)

Mor

talit

yO

nset

of P

LI

No

PLI

PLI

Inci

denc

e**N

o PL

IPL

Ip

valu

eN

o PL

IPL

Ip

valu

e

All p

atien

ts31

50 (9

6.1)

123

(3.9

)0.

991.

1 ±

3.7

8.0

± 12

.4<

0.00

145

(1.4

)25

(20.

3)<

0.00

12.

9 ±

2.2

Conv

entio

nal

venti

latio

n10

41 (9

3.3)

75 (6

.7)

1.71

1.0

± 3.

09.

2 ±

10.2

< 0.

001

17 (1

.4)

17 (2

0.7)

< 0.

001

2.5

± 1.

5

Prot

ectiv

e ve

ntila

tion

1762

(98.

0)37

(2.0

)0.

461.

3 ±

4.3

6.1

± 16

.2<

0.00

128

(1.5

)8

(20.

0)<

0.00

13.

9 ±

3.3

Age

<

65 y

ears

65 y

ears

1504

(9.2

)13

90 (9

5.3)

44 (2

.8)

68 (4

.7)

0.76

1.02

0.8

± 2.

61.

5 ±

4.7

5.0

± 6.

010

.5 ±

15.

4<

0.00

1<

0.00

18

(0.5

)37

(2.5

)6

(12.

8)19

(25.

3)<

0.00

1<

0.00

12.

5 ±

1.2

3.2

± 2.

6

ASA

scor

e

< 3

313

91 (9

6.6)

1106

(95.

3)49

(3.4

)55

(4.7

)0.

811.

141.

0 ±

4.0

1.8

± 4.

44.

2 ±

5.3

9.0

± 14

.40.

001

0.01

710

(0.7

)25

(2.3

)9

(17.

0)16

(29.

1)<

0.00

1<

0.00

12.

5 ±

1.2

3.4

± 2.

9

Surg

ery

Ab

dom

inal

Th

orac

ic17

98 (9

6.6)

1285

(95.

7)64

(3.4

)58

(4.3

)0.

791.

321.

0 ±

4.1

1.5

± 0.

99.

0 ±

14.5

5.9

± 2.

1<

0.00

1<

0.00

132

(1.8

)13

(1.0

)9

(14.

1)16

(27.

6)<

0.00

1<

0.00

12.

3 ±

0.9

3.4

± 2.

7

PLI:

post

oper

ative

lung

inju

ry; I

CU: i

nten

sive

care

uni

t; LO

S: le

ngth

of s

tay;

Ons

et o

f PLI

is e

xpre

ssed

in d

ays

* In

som

e ca

ses t

he n

umbe

r of p

atien

ts a

re n

ot a

ddin

g up

due

to m

issin

g va

lues

** E

xpre

ssed

as c

ases

per

per

son-

year

IPD metaanalysis postoperative lung injury

Chap

ter

10

233

13. There was no interaction between mortality and the design of the study (RCT versus No-RCT) in the overall cohort (p for interaction = 0.11), abdominal surgery (p for interaction = 0.23), or thoracic surgery (p for interaction = 0.78) (Appendix figure 14).

Based on the excluded studies where the data is available,21,23,24 the crude incidence of PLI was 6.6%, similar to that found in our study (p = 0.116). Also, protective ventilation was associated with lower incidence of PLI compared to conventional ventilation (5.3% vs. 13.1%; p = 0.031), similar to the findings of the present study. There was no interaction between mortality and PLI incidence and the excluded analysis (Appendix figure 15).

Discussion

This individual patient data metaanalysis shows that development of postoperative lung injury is associated with high attributable mortality. In addition, development of postoperative lung injury is associated with an important increase in resource use as reflected in longer ICU and hospital length of stay. The incidence of postoperative lung injury is similar in patients undergoing abdominal or thoracic surgery. However, the attributable mortality of this condition is higher in those submitted to thoracic procedures. Intraoperative protective ventilation is associated with lower incidence of postoperative lung injury compared to conventional ventilation in abdominal or thoracic surgery. However, if postoperative lung injury develops, intraoperative protective strategy of ventilation is not associated with reduced attributable mortality, at least suggesting that the benefits of this strategy of ventilation is mainly due the reduction in the incidence of postoperative lung injury.

Table 4. Outcome of patients with postoperative lung injury

Variable HR of Mortality (95% CI)

HR of ICU Discharge (95% CI)

All patients 9.58 (5.32 – 17.34) 0.45 (0.33 – 0.66)

Conventional ventilation 14.22 (5.91 – 34.26) 0.39 (0.25 – 0.58)

Protective ventilation 6.07 (2.47 – 14.55) 0.71 (0.42 – 1.19)

Age < 65 years ≥ 65 years

33.10 (8.32 – 131.39)7.32 (3.72 – 14.23)

0.52 (0.32 – 0.83)0.43 (0.26 – 0.68)

ASA score < 3 ≥ 3

26.67 (9.44 – 75.22)6.05 (2.91 – 12.66)

0.55 (0.34 – 0.92)0.41 (0.26 – 0.60)

Surgery Abdominal Thoracic

7.12 (2.67 – 19.08)10.46 (4.72 – 23.18)

0.47 (0.32 – 0.69)0.19 (0.08 – 0.40)

HR: hazard ratio; ICU: intensive care unit; CI: confidence interval

234

Our analysis presents several strengths. First, we performed an individual patient data metaanalysis including a larger number of patients with a higher number of variables and well–documented risk factors compared to previous studies.1-4 Second, the primary and secondary outcomes were clearly defined in the majority of the investigations and the definition of postoperative lung injury was done a priori. Third, we included the two most recent largest and well–performed randomized controlled trials in the field.18,19 Fourth, we analysed patients undergoing abdominal and thoracic surgery and they are at high risk for development of postoperative lung injury.1 Finally, individual patient data metaanalysis including a higher number of randomized controlled trials have some advantages compared to large observational studies.

Since the number of major surgical procedures undertaken worldwide each year is high, the finding that postoperative lung injury is associated with such a bad outcome is important. Identification of mechanisms contributing to the development of this complication and finding of preventive measures is highly needed. The use of lower tidal volumes during intraoperative ventilation appears to be a clinically relevant and modifiable strategy.2,18 At the same time, ventilation strategies that use high PEEP levels are associated with potentially dangerous side–effects, as reported in the last randomized controlled trial comparing high with low PEEP levels in patients under intraoperative ventilation with low tidal volume.19

The results of this metaanalysis are, at least in part in line with results from previous investigations. Studies suggest that in patients undergoing high risk elective surgery,2 the reported incidence of postoperative lung injury was 3%, similar to what the present metaanalysis found.

Figure 3. Kaplan–Meier estimates of the probability of overall survivalData for the Kaplan–Meier estimates of the probability of overall survival in: A) patients undergoing thoracic surgery without postoperative lung injury (black solid line), patients undergoing thoracic surgery with postoperative lung injury (black dotted line), patients undergoing abdominal surgery without postoperative lung injury (gray solid line), and patients undergoing abdominal surgery with postoperative lung injury (gray dotted line); and in B) patients undergoing protective ventilation without postoperative lung injury (black solid line), patients undergoing protective ventilation with postoperative lung injury (black dotted line), patients undergoing conventional ventilation without postoperative lung injury (gray solid line), and patients undergoing conventional ventilation with postoperative lung injury (gray dotted line). Data were censored at 30 days after surgery. Abd: abdominal; Tho: thoracic; LI: lung injury; Pro: protective; Conv: conventional

IPD metaanalysis postoperative lung injury

Chap

ter

10

235

That same investigation showed a similar increased length of hospital stay in patients who developed postoperative lung injury, and an increased in–hospital mortality of as high as 17%. Furthermore, in that study 60–day and one–year survival of patients with postoperative lung injury was lower than that of patients who did not develop this complication. Studies in other patient groups show similar findings.12,22 It is important to emphasize that incidence and outcomes of postoperative lung injury might be different in different types of surgery3 and according to definitions used.

The findings of this metaanalysis add to our knowledge on the outcome of postoperative lung injury, and the potential measures to prevent this important complication after major surgery, mainly the use of protective strategies of mechanical ventilation during surgery. This is of particular importance for those who apply intraoperative ventilation, since most of the time they follow on patients’ outcomes only the first postoperative day. Indeed, the metaanalysis shows that many patients do develop lung injury after major surgery beyond the first postoperative day. Second, development of postoperative lung injury was dependent on the intraoperative ventilation strategy, and thus should be seen as a potentially preventable complication. Finally, the attributable mortality of postoperative lung injury after intraoperative ventilation using conventional settings is similar to those after lung–protective ventilation, meaning that the benefits of intraoperative protective ventilation is mainly due the reduction of the incidence of postoperative lung injury. This finding might simply reflects the heterogeneous factors that cause death in this population well beyond just postoperative lung injury.

The finding that patients who received conventional intra–operative ventilation and developed postoperative lung injury died earlier than those who developed this complication after protective intraoperative ventilation, even though attributable mortality at the end of follow up was the same, is intriguing. One possible explanation is that patients who did not receive intraoperative lung–protective ventilation could have been ventilated with injurious ventilation after surgery, and maybe even after development of lung injury. Unfortunately, it was impossible to collect data on ventilator settings after surgery, therefore our reasoning, although plausible, remains speculative.

The observed incidence and outcome of postoperative lung injury might not be easily appreciated during daily clinical work among physicians, surgeons, and anaesthesiologists involved in the perioperative management. In fact, this is a large cohort evaluating the development and impact of postoperative lung injury in patients undergoing elective abdominal and thoracic procedures. The finding that postoperative lung injury develops after the first postoperative day is important, suggesting we need a more precise timing for monitoring and clinical management.

Some limitations should also be discussed. First, our analyses were restricted to studies of intraoperative protective ventilation and data on postoperative ventilation are not available. Second, not all investigators could provide individual patient data, and, therefore, data from seven studies were not included. However, the results of a classical metaanalysis are in agreement with those found in the present analysis.4 Third, there was no information on choices of treatment after surgery, like postoperative ventilation, blood transfusion policies, fluid regimens, pain control, cardiac protection and others, and it could be anaesthetists that applied protective

236

ventilation also provided other lung protective strategies after surgery. Therefore, we cannot answer the question of how much the in–hospital mortality is influenced by the adequacy of treatment. Also, we do not know the reason of death in each patient. Fourth, since there is no gold standard for the diagnosis of postoperative lung injury, misclassification of patients with this complication might have underestimated as well as overestimated the observed attributable morbidity. Fifth, we were not able to collect information regarding history of previous abdominal or thoracic surgery, which can influence the outcome. Sixth, these results should be analysed within the context of the included studies and one limitation is that we pooled data from studies with heterogeneous research methodologies and with significant quantitative heterogeneity. Seventh, the number of studies included is moderate and therefore the model may lack power to detect association and are unable to ascertain multiple potential sources of confounding. Eight, we did not include previous pulmonary alterations as co-variate in the model however, only 4.9% of the population analysed presented chronic obstructive pulmonary disease (COPD) or other chronic pulmonary disease. Ninth, it should be noted that minimal invasive techniques for thoracic surgery are increasingly used, and these techniques usually last shorter and are associated with a shorter length of stay. Since the studies included in the present analysis did not include these techniques, we cannot assess the outcomes and effects of protective ventilation during minimal invasive procedures. However, a recent study suggests that protective ventilation strategies benefits patients undergoing minimally invasive esophagectomy as well.32 Finally, although the type of intraoperative ventilation appears to play an important role in development of acute lung injury, other factors during postoperative management may plausibly be contributory, especially for patients who developed lung injury days after surgery (or cessation of mechanical ventilation).

In conclusion, based on an individual patient data from seven studies of intraoperative protective ventilation, development of postoperative lung injury is associated with high attributable mortality. The attributable mortality of postoperative lung injury is higher in patients undergoing thoracic compared to abdominal surgery. Intraoperative protective ventilation is associated with lower incidence of postoperative lung injury, but seems not to affect in–hospital mortality.

Role of the funding sourceThe corresponding author had full access to all of the data and the final responsibility to submit for publication.

IPD metaanalysis postoperative lung injury

Chap

ter

10

237

Research in context

Systematic reviewWe searched MEDLINE, Cumulative Index to Nursing and Allied Health Literature (CINAHL), Web of Science, and Cochrane Central Register of Controlled Trials (CENTRAL) up to April 2014. The sensitive search strategy combined the following Medical Subject Headings and Keywords ([protective ventilation OR lower tidal volume OR low tidal volume OR positive end–expiratory pressure OR positive end expiratory pressure OR PEEP]). We restricted our analysis to studies in surgery. We assessed the quality of identified studies to ensure minimization of bias.

InterpretationDevelopment of postoperative lung injury is associated with high attributable mortality. The attributable mortality of postoperative lung injury is higher in patients undergoing thoracic compared to abdominal surgery. Intraoperative protective ventilation is associated with lower incidence of postoperative lung injury, but seems not to affect in–hospital mortality.

238

References1. Canet J, Gallart L, Gomar C, et al. Prediction of postoperative pulmonary complications in a population-based surgical

cohort. Anesthesiology 2010; 113:1338-502. 2Hemmes SN, Serpa Neto A, Schultz MJ. Intraoperative ventilatory strategies to prevent postoperative pulmonary

complications: a metaanalysis. Curr Opin Anaesthesiol 2013; 26:126-333. Futier E, Constantin J, Jaber S. Protective lung ventilation in operating room: Systematic Review. Minerva Anestesiol

2014; 80:726-354. Serpa Neto A, Cardoso SO, Manetta JA, et al. Association between use of lung-protective ventilation with lower

tidal volumes and clinical outcomes among patients without acute respiratory distress syndrome: a metaanalysis. JAMA 2012; 308:1651-9

5. Levin MA, McCormick PJ, Lin HM, Hosseinian L, Fischer GW. Low intraoperative tidal volume ventilation with minimal PEEP is associated with increased mortality. Br J Anaesth 2014;113:97-108

6. Serpa Neto A, Hemmes SNT, Gama de Abreu M, Pelosi P, Schultz MJ; Protective Ventilation Network (PROVENet). Protocol for a systematic review and individual patient metaanalysis of benefit of so-called lung-protective ventilation-settings in patients under general anesthesia for surgery. Syst Rev 2014; 3:2-6

7. Bernard GR, Artigas A, Brigham KL, et al. The American-European Consensus Conference on ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination. Am J Respir Crit Care Med 1994; 149:818-24

8. Wrigge H, Uhlig U, Zinserling J, et al. The effects of different ventilatory settings on pulmonary and systemic inflammatory responses during major surgery. Anesth Analg 2004; 98:775-81

9. Schilling T, Kozian A, Huth C, et al. The pulmonary immune effects of mechanical ventilation in patients undergoing thoracic surgery. Anesth Analg 2005; 101:957-65

10. Wolthuis EK, Choi G, Dessing MC, et al. Mechanical ventilation with lower tidal volumes and positive end-expiratory pressure prevents pulmonary inflammation in patients without preexisting lung injury. Anesthesiology 2008; 108:46-54

11. Lin WQ, Lu XY, Cao LH, Wen LL, Bai XH, Zhong ZJ. Effects of the lung protective ventilatory strategy on proinflammatory cytokine release during one-lung ventilation. Ai Zheng 2008; 27:870-3

12. Licker M, Diaper J, Villiger Y, et al. Impact of intraoperative lung-protective interventions in patients undergoing lung cancer surgery. Crit Care 2009; 13:R41-50

13. Weingarten TN, Whalen FX, Warner DO, et al. Comparison of two ventilatory strategies in elderly patients undergoing major abdominal surgery. Br J Anaesth 2010; 104:16-22

14. Fernandez-Bustamante A, Wood CL, Tran ZV, Moine P. Intraoperative ventilation: incidence and risk factors for receiving large tidal volumes during general anesthesia. BMC Anesthesiol 2011; 11:22-9

15. Treschan TA, Kaisers W, Schaefer MS, et al. Ventilation with low tidal volumes during upper abdominal surgery does not improve postoperative lung function. Br J Anaesth 2012; 109:263-71

16. Unzueta C, Tusman G, Suarez-Sipmann F, Böhm S, Moral V. Alveolar recruitment improves ventilation during thoracic surgery: a randomized controlled trial. Br J Anaesth 2012; 108:517-24

17. Severgnini P, Selmo G, Lanza C, et al. Protective mechanical ventilation during general anesthesia for open abdominal surgery improves postoperative pulmonary function. Anesthesiology 2013; 118:1307-21

18. Futier E, Constantin JM, Paugam-Burtz C, et al. A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. NEJM 2013; 369:428-37

19. The PROVE Network Investigators. Higher versus lower positive end-expiratory pressure during general anaesthesia for open abdominal surgery – The PROVHILO trial. Lancet 2014;384:495-503

20. Chaney MA, Nikolov MP, Blakeman BP, Bakhos M. Protective ventilation attenuates postoperative pulmonary dysfunction in patients undergoing cardiopulmonary bypass. J Cardiothorac Vasc Anesth 2000; 14:514-8

21. Michelet P, D’Journo XB, Roch A, et al. Protective ventilation influences systemic inflammation after esophagectomy: a randomized controlled study. Anesthesiology 2006; 105:911-9

22. Cai H, Gong H, Zhang L, Wang Y, Tian Y. Effect of low tidal volume ventilation on atelectasis in patients during general anesthesia: a computed tomographic scan. J Clin Anesth 2007; 19:125-9

23. Yang M, Ahn HJ, Kim K, et al. Does a protective ventilation strategy reduce the risk of pulmonary complications after lung cancer surgery?: a randomized controlled trial. Chest 2011; 139:530-7

24. Ahn HJ, Kim JA, Yang M, Shim WS, Park KJ, Lee JJ. Comparison between conventional and protective one-lung ventilation for ventilator-assisted thoracic surgery. Anaesth Intensive Care 2012; 40:780-8

25. Lellouche F, Dionne S, Simard S, Bussières J, Dagenais F. High tidal volumes in mechanically ventilated patients increase organ dysfunction after cardiac surgery. Anesthesiology 2012; 116:1072-82

26. Koner O, Celebi S, Balci H, Cetin G, Karaoglu K, Cakar N. Effects of protective and conventional mechanical ventilation on pulmonary function and systemic cytokine release after cardiopulmonary bypass. Intensive Care Med 2004; 30:620-6

27. Wrigge H, Uhlig U, Baumgarten G, et al. Mechanical ventilation strategies and inflammatory responses to cardiac

IPD metaanalysis postoperative lung injury

Chap

ter

10

239

surgery: a prospective randomized clinical trial. Intensive Care Med 2005; 31:1379-8728. Zupancich E, Paparella D, Turani F, et al. Mechanical ventilation affects inflammatory mediators in patients undergoing

cardiopulmonary bypass for cardiac surgery: A randomized clinical trial. J Thorac Cardiovasc Surg 2005; 130:378-8329. Reis Miranda D, Gommers D, Struijs A, et al. Ventilation according to the open lung concept attenuates pulmonary

inflammatory response in cardiac surgery. Eur J Cardiothorac Surg 2005; 28:889-9530. Sundar S, Novack V, Jervis K, et al. Influence of low tidal volume ventilation on time to extubation in cardiac surgical

patients. Anesthesiology 2011; 114:1102–1031. Memtsoudis SG, Bombardieri AM, Ma Y, Girardi FP. The effect of low versus high tidal volume ventilation on

inflammatory markers in healthy individuals undergoing posterior spine fusion in the prone position: a randomized controlled trial. J Clin Anesth 2012; 24:263-9

32. Shen Y, Zhong M, Wu W, et al. The impact of tidal volume on pulmonary complications following minimally invasive esophagectomy: a randomized and controlled study. J Thorac Cardiovasc Surg 2013;146:1267-7

240

Supplementary Appendix to: ‘Incidence of mortality and morbidities of postoperative lung injury in abdominal and thoracic surgery patients: A systematic review and metaanalysis

Ary Serpa Neto MD MSc, Sabrine NT Hemmes MD, Carmen SV Barbas MD PhD, Martin Beiderlinden MD, Michelle Biehl MD, Ana Fernandez-Bustamante MD PhD, Emmanuel Futier MD PhD, Ognjen Gajic MD PhD, Samir Jaber MD PhD, Alf Kozian MD PhD, Marc Licker MD, Wen-Qian Lin MD, Stavros G Memtsoudis MD PhD, Dinis Reis Miranda MD, Pierre Moine MD, Domenico Paparella MD, Marco Ranieri MD PhD, Federica Scavonetto MD, Thomas Schilling MD PhD DEEA, Gabriele Selmo MD, Paolo Severgnini MD, Juraj Sprung MD PhD, Sugantha Sundar MD, Daniel Talmor MD PhD, Tanja Treschan MD, Gerardo Tusman MD PhD, Maria Carmen Unzueta MD, Toby N Weingarten MD, Esther K Wolthuis MD PhD, Hermann Wrigge MD PhD, Marcelo Gama de Abreu MD PhD, Paolo Pelosi MD, Marcus J Schultz MD PhD

eFigure 1. Literature search strategy

IPD metaanalysis postoperative lung injury

Chap

ter

10

241

eFigure 2. Individual and pooled incidence rate of postoperative lung injury

eFigure 3. In-hospital mortality in patients who developed or not postoperative lung injury in thoracic or abdominal surgery

242

eFigure 4. Interaction between mortality and design of the study

eFigure 5. Interaction between outcome and excluded studies

IPD metaanalysis postoperative lung injury

Chap

ter

10

243

eTable 1. Characteristics of included studies

Study, year Allocation Concealment Baseline Similarity Early

StoppingaLost to Follow-up

Intention-to-Treat Analysis

Wrigge, 2004 Sealed envelopes

Age: similarIllness severity: similar (ASA)

No 4.6% NS

Schilling, 2005 Random numbers Age: similar No No NS

Wolthuis, 2008 Sealed envelopes

Age: similarIllness severity: similar (ASA)

No No No

Lin, 2008 NS Age: similar No No NS

Licker, 2009 Not applicable

Age: similarIllness severity: favour higher VT (ASA)

Not applicable

Not applicable Not applicable

Fernandez-Bustamante, 2011 Not applicable

Age: similarIllness severity: similar (ASA)

Not applicable

Not applicable Not applicable

Weingarten, 2010 ScheduleAge: similarIllness severity: similar (ASA)

No No NS

Treschan, 2012 Sealed envelopes

Age: similarIllness severity: similar (ASA)

No No Yes

Unzueta, 2012 Random tableAge: similarIllness severity: similar (ASA)

No No NS

Severgnini, 2013 Sealed envelopes

Age: similarIllness severity: similar (ASA)

No 1.7% NS

Futier, 2013 Computer-Generated

Age: similarIllness severity: similar (PORI)

No No Yes

Hemmes, 2014 Computer-Generated

Age: similarIllness severity: similar (ARISCAT)

No No Yes

NS: not specified; VT: tidal volume; STSMS: society of thoracic surgeons mortality score; PORI: preoperative risk indexa: Early termination for benefit or futility and the presence of an explicit a priori stopping rules

244

eTable 2. Jadad scale

StudiesWas the study described as randomized?

Was the study described as double blind?

Was there a description of withdrawals and dropouts?

The method of randomization was described in the paper, and that method was appropriate.

The method of blinding was described, and it was appropriate.

Wrigge, 2004 Yes No Yes No No

Schilling, 2005 Yes No Yes No No

Wolthuis, 2008 Yes No Yes Yes No

Lin, 2008 Yes No No No No

Weingarten, 2010 Yes No Yes Yes No

Treschan, 2012 Yes Yes Yes Yes Yes

Unzueta, 2012 Yes No Yes No No

Severgnini, 2013 Yes Yes Yes Yes Yes

Futier, 2013 Yes Yes Yes Yes Yes

Hemmes, 2014 Yes Yes Yes Yes Yes

eTable 3. GRACE checklist1

Studies

Adequate Treatment

Adequate Outcomes

Objective Outcomes

Valid Outcomes

Similar Outcomes

Covariates Recorded New Initiators Concurrent

ComparatorsCovariates Accounted For

Immortal Time Bias

Sensitivity Analysis

D1 D2 D3 D4 D5 D6 M1 M2 M3 M4 M5

Licker, 2009 Yes Yes Yes Yes Yes Yes Yes Yes Yes Yes Yes

Fernandez, 2011 Yes Yes Yes Yes Yes Yes Yes Yes Yes Yes Yes1Dreyer NA, Velentgas P, Westrich K, Dubois R. The GRACE checklist for rating the quality of observational studies of comparative effectiveness: a tale of hope and caution. J Manag Care Pharm 2014; 20: 301-8

IPD metaanalysis postoperative lung injury

Chap

ter

10

245

eTable 4. Demographics of patients undergoing thoracic surgery

Variable Total(n = 1374)

No PLI(n = 1285)

PLI(n = 58) p value

Age, years 61.73 ± 11.85 61.57 ± 11.88 66.00 ± 10.35 0.005

Gender, male 872 (63.4) 837 (65.1) 35 (60.3) 0.482

ASA 2.45 ± 0.63 2.44 ± 0.63 2.72 ± 0.64 0.001

BMI, kg/m2 25.10 ± 4.43 25.10 ± 4.45 25.07 ± 4.36 0.956

Predisposing conditions Shock Pneumonia Transfusion of blood products Sepsis

1 (0.0)9 (0.6)39 (2.8)5 (0.3)

0 (0.0)4 (0.3)26 (2.0)1 (0.0)

1 (1.7)5 (8.6)13 (22.4)4 (6.8)

0.714< 0.001< 0.001< 0.001

Ventilatory Parameters* Tidal volume, ml/kg PBW Respiratory rate, mpm FiO2, %

7.78 ± 2.0512.90 ± 2.2145.00 ± 16.61

7.64 ± 1.8412.94 ± 2.2145.17 ± 16.86

9.54 ± 2.5311.81 ± 1.8940.40 ± 5.07

< 0.0010.0010.064

Oxygenation Parameters* pH PaO2 / FiO2

PaCO2

7.26 ± 0.12392.64 ± 118.4541.17 ± 6.89

7.28 ± 0.10398.87 ± 118.6941.13 ± 7.05

7.13 ± 0.15294.28 ± 55.2241.86 ± 3.31

< 0.0010.0010.689

ICU LOS, days 1.88 ± 1.52 1.57 ± 0.90 5.93 ± 2.15 < 0.001

Hospital LOS, days 13.05 ± 6.47 12.71 ± 6.10 20.72 ± 9.14 < 0.001

In-Hospital Mortality, % 29 (2.1) 13 (1.0) 16 (27.5) < 0.001

PLI: postoperative lung injury; BMI: body mass index; PBW: predicted body weight; MPM: movements per minute; FiO2: inspired fraction of oxygen; ICU: intensive care unit; LOS: length of stay; *: in the end of the surgery

246

eTable 5. Demographics of patients undergoing abdominal surgery

Variable Total(n = 1991)

No PLI(n = 1798)

PLI(n = 64) p value

Age, years 63.24 ± 13.25 63.20 ± 13.21 66.75 ± 12.66 0.035

Gender, male 1235 (62.0) 1191 (66.2) 44 (68.7) 0.433

ASA 2.32 ± 0.67 2.34 ± 0.67 2.43 ± 0.63 0.347

BMI, kg/m2 26.13 ± 5.02 26.11 ± 5.09 27.12 ± 5.00 0.122

Predisposing conditions Shock Pneumonia Transfusion of blood products Sepsis

51 (2.5)8 (0.4)157 (7.8)7 (0.3)

50 (2.7)7 (0.3)144 (8.0)7 (0.3)

1 (1.5)1 (1.5)13 (20.3)0 (0.0)

0.7140.231< 0.0010.871

Ventilatory Parameters* Tidal volume, ml/kg PBW Respiratory rate, mpm FiO2, %

8.56 ± 1.8011.00 ± 2.8644.87 ± 9.64

8.53 ± 1.7610.97 ± 2.8345.34 ± 10.07

9.15 ± 1.7211.62 ± 2.3840.78 ± 2.70

0.0060.0930.011

Oxygenation Parameters* pH PaO2 / FiO2

PaCO2

7.35 ± 0.06334.73 ± 220.7638.62 ± 5.16

7.33 ± 0.05365.58 ± 230.3138.49 ± 3.79

7.43 ± 0.04308.14 ± 214.232.42 ± 4.62

0.4620.2450.493

ICU LOS, days 1.41 ± 5.24 1.08 ± 4.10 9.00 ± 14.57 < 0.001

Hospital LOS, days 16.53 ± 18.45 16.17 ± 17.92 21.09 ± 23.58 0.035

In-Hospital Mortality, % 42 (2.1) 33 (1.8) 9 (14.1) < 0.001

PLI: postoperative lung injury; BMI: body mass index; PBW: predicted body weight; MPM: movements per minute; FiO2: inspired fraction of oxygen; ICU: intensive care unit; LOS: length of stay; *: in the end of the surgery

eTable 6. Ventilatory parameters and duration of ventilation

Parameters

Total Abdominal Thoracic

Protective(n = 1799)

Conventional(n = 1116) p value Protective

(n = 1155)Conventional(n = 707) p value Protective

(n = 707)Conventional(n = 636) p-value

Tidal volume, ml/kg PBW 7.10 ± 1.15 9.95 ± 1.62 < 0.01 7.54 ± 0.93 10.40 ± 1.52 < 0.01 6.34 ± 1.09 9.46 ± 1.58 < 0.01

PEEP, cmH2O 5.41 ± 4.10 2.79 ± 2.68 < 0.01 6.44 ± 4.57 2.72 ± 3.29 < 0.01 3.61 ± 2.08 2.87 ± 1.77 < 0.01

Respiratory rate, bpm 12.91 ± 2.82 10.45 ± 1.77 < 0.01 11.76 ± 2.97 9.60 ± 1.97 < 0.01 14.38 ± 1.73 11.15 ± 1.21 < 0.01

Plateau pressure, cmH2O 16.52 ± 5.87 16.49 ± 4.03 0.912 19.12 ± 5.81 17.32 ± 6.53 < 0.01 13.63 ± 4.43 16.23 ± 2.75 < 0.01

Duration of ventilation, minutes 369.23 ± 758.59 408.53 ± 844.14 0.857 382.74 ± 788.87 487.21 ± 755.43 0.101 401.12 ± 672.21 498.21 ± 512.32 0.222

ICU length of stay, days 1.49 ± 5.24 1.51 ± 4.33 0.935 1.42 ± 5.92 1.40 ± 4.59 0.958 1.71 ± 0.99 2.13 ± 2.04 0.045

Hospital length of stay, days 15.77 ± 16.75 14.01 ± 14.97 0.231 15.99 ± 17.34 15.76 ± 13.76 0.121 12.01 ± 5.80 14.28 ± 6.98 < 0.01

PBW: predicted body weight; PEEP: positive end-expiratory pressure; BPM: beats per minute; ICU: intensive care unit

IPD metaanalysis postoperative lung injury

Chap

ter

10

247

eTable 7. Incidence of postoperative lung injury and its characteristics in patients undergoing thoracic surgery

Postoperative Lung Injury No-Postoperative Lung Injury

Number of Patients Onset (days) Mortality ICU LOS

(days)Number of Patients Mortality ICU LOS

(days)

All patients 58 (4.3) 3.4 ± 2.7 16 (27.6) 5.9 ± 2.1 1285 (95.7) 13 (1.0) 1.5 ± 0.9

Conventional ventilation 42 (6.7) 2.7 ± 1.7 12 (28.6) 6.0 ± 2.1 578 (93.3) 4 (0.7) 1.3 ± 0.7

Protective ventilation 16 (2.2) 5.5 ± 4.2 4 (25.0) 4.0 ± 4.5 707 (97.8) 9 (1.3) 1.6 ± 0.9

Age < 65 years ≥ 65 years

25 (3.3)33 (5.5)

2.6 ± 1.33.9 ± 3.3

6 (24.0)10 (30.3)

5.1 ± 1.16.8 ± 2.7

717 (96.7)566 (94.5)

1 (0.1)12 (2.1)

1.4 ± 0.81.7 ± 0.9

ASA score < 3 ≥ 3

22 (3.0)36 (5.8)

2.9 ± 1.43.6 ± 3.1

8 (36.4)8 (22.2)

5.5 ± 2.06.4 ± 2.3

694 (97.0)581 (94.2)

3 (0.4)10 (1.7)

1.4 ± 0.81.6 ± 0.9

ICU: intensive care unit; LOS: length of stay

248

eTable 8. Incidence of postoperative lung injury and its characteristics in patients undergoing abdominal surgery

Postoperative Lung Injury No-Postoperative Lung Injury

Number of Patients

Onset (days) Mortality ICU LOS

(days)Number of Patients Mortality ICU LOS

(days)

All patients 64 (3.4) 2.3 ± 0.9 9 (14.1) 9.0 ± 14.5 1798 (96.6) 32 (1.8) 1.0 ± 4.1

Conventional ventilation 40 (6.0) 2.2 ± 1.0 5 (12.5) 11.1 ± 12.6 620 (94.0) 13 (2.1) 0.9 ± 3.2

Protective ventilation 24 (1.9) 2.4 ± 0.8 4 (16.7) 6.2 ± 16.7 1178 (98.1) 19 (1.6) 1.2 ± 4.9

Age < 65 years ≥ 65 years

22 (2.4)42 (4.3)

2.2 ± 0.92.3 ± 1.0

0 (0.0)9 (21.4)

4.9 ± 7.411.6 ± 17.3

884 (97.6)914 (95.7)

7 (0.8)25 (2.7)

0.6 ± 2.91.5 ± 5.1

ASA score < 3 ≥ 3

31 (3.7)24 (3.9)

2.2 ± 1.02.5 ± 0.5

1 (3.2)5 (20.8)

3.5 ± 6.510.0 ± 16.8

797 (96.3)577 (96.1)

7 (0.9)14 (2.4)

0.9 ± 4.71.8 ± 5.1

ICU: intensive care unit; LOS: length of stay

eTable 9. Characteristics of patients with postoperative lung injury

Variable Total(n = 123)

Survivors(n = 98)

Non-Survivors(n = 25) p value

Age, years 66.39 ± 11.58 65.14 ± 11.96 71.24 ± 8.52 0.018

Gender, male 78 (63.4) 61 (62.2) 17 (68.0) 0.635

ASA 2.58 ± 0.65 2.54 ± 0.63 2.72 ± 0.70 0.252

BMI, kg/m2 26.14 ± 4.79 26.24 ± 4.92 25.74 ± 4.36 0.643

Ventilatory Parameters* Tidal volume, ml/kg PBW PEEP, cmH2O Respiratory rate, mpm FiO2, %

9.34 ± 2.142.86 ± 3.4311.71 ± 2.1740.56 ± 4.21

9.33 ± 2.122.88 ± 3.6011.40 ± 2.2655.55 ± 22.36

9.36 ± 2.292.80 ± 2.7010.66 ± 1.6357.11 ± 25.02

0.9430.9110.4500.845

Duration of ventilation, hours 30.35 ± 75.50 29.80 ± 78.95 34.13 ± 47.81 0.867

ICU LOS, days 8.17 ± 12.55 8.34 ± 13.23 7.00 ± 6.37 0.793

Hospital LOS, days 20.91 ± 18.08 20.33 ± 17.42 23.16 ± 20.66 0.489

BMI: body mass index; PBW: predicted body weight; MPM: movements per minute; FiO2: inspired fraction of oxygen; ICU: intensive care unit; LOS: length of stay; PEEP: positive-end expiratory pressure; *: in the middle of the surgery

IPD metaanalysis postoperative lung injury

Chap

ter

10

249

eTab

le 1

0. C

hara

cter

istic

s of

the

patie

nts

with

pos

tope

rativ

e lu

ng in

jury

und

er p

rote

ctive

or c

onve

ntion

al v

entil

ation

Varia

bles

Gen

eral

(n =

122

)

p-va

lue

Thor

acic

(n =

58)

p-va

lue

Abdo

min

al (n

= 6

4)

p-va

lue

Prot

ectiv

e (n

= 4

0)Co

nven

tiona

l (n

= 8

2)Pr

otec

tive

(n =

16)

Conv

entio

nal

(n =

42)

Prot

ectiv

e (n

= 2

4)Co

nven

tiona

l (n

= 4

0)

Age,

yea

rs64

.3 ±

14.

067

.4 ±

10.

20.

169

63.2

± 1

4.3

67.0

± 8

.40.

215

65.0

± 1

4.0

67.8

± 1

1.9

0.40

7

Gen

der,

mal

e28

(70.

0)50

(61.

0)0.

330

ASA

2.58

± 0

.72.

58 ±

0.6

0.94

72.

8 ±

0.5

2.7

± 0.

70.

523

2.4

± 0.

72.

4 ±

0.6

0.98

8

BMI,

kg/m

225

.7 ±

5.3

26.3

± 4

.50.

496

25.4

± 5

.525

.0 ±

3.9

0.75

226

.0 ±

5.3

27.9

± 4

.70.

144

Venti

lato

ry P

aram

eter

s*

Ti

dal v

olum

e, m

l/kg

PBW

PE

EP, c

mH 2

O

Resp

irato

ry ra

te, m

pm

FiO

2, %

7.4

± 1.

35.

7 ±

3.9

13.1

± 2

.440

.0 ±

4.4

10.3

± 1

.81.

5 ±

2.1

10.8

± 1

.441

.0 ±

4.1

< 0.

001

< 0.

001

< 0.

001

0.39

6

7.2

± 1.

92.

6 ±

1.5

13.5

± 1

.638

.5 ±

4.6

10.4

± 2

.11.

3 ±

1.6

10.9

± 1

.341

.4 ±

5.0

< 0.

001

0.00

8<

0.00

10.

068

7.5

± 0.

67.

8 ±

3.6

12.9

± 2

.855

.6 ±

22.

1

10.2

± 1

.31.

6 ±

2.5

10.7

± 1

.555

.8 ±

23.

1

< 0.

001

< 0.

001

< 0.

001

0.96

8

Oxy

gena

tion

Para

met

ers*

pH

a

PaO

2 / F

iO2

Pa

CO2

7.32

± 0

.11

258.

5 ±

103.

735

.5 ±

3.5

7.14

± 0

.16

317.

0 ±

181.

142

.1 ±

3.1

5

0.16

50.

367

0.20

4

--- --- ---

--- --- ---

--- --- ---

7.38

± 0

.01

247.

1 ±

104.

637

.1 ±

0.2

7.45

± 0

.03

345.

7 ±

257.

130

.5 ±

4.0

0.49

50.

318

0.08

1

Dura

tion

of v

entil

ation

, ho

urs

28.8

± 7

0.3

31.1

± 7

8.4

0.90

4---

---29

.9 ±

71.

640

.4 ±

89.

50.

626

ICU

LO

S, d

ays

6.1

± 16

.29.

2 ±

10.3

0.39

3---

---6.

3 ±

16.7

11.1

± 1

2.6

0.29

6

Hosp

ital L

OS,

day

s21

.2 ±

24.

120

.8 ±

14.

60.

905

20.1

± 5

.421

.0 ±

10.

20.

737

22.0

± 3

1.3

20.6

± 1

8.2

0.82

0

In-H

ospi

tal M

orta

lity,

%8

(20.

0)17

(20.

7)0.

925

4 (2

5.0)

12 (2

8.6)

0.78

64

(16.

7)5

(12.

5)0.

642

BMI:

body

mas

s ind

ex; P

BW: p

redi

cted

bod

y w

eigh

t; M

PM: m

ovem

ents

per

min

ute;

FiO

2: in

spire

d fr

actio

n of

oxy

gen;

ICU

: int

ensiv

e ca

re u

nit;

LOS:

leng

th o

f sta

y; P

EEP:

pos

itive

-end

ex

pira

tory

pre

ssur

e; *

: in

the

mid

dle

of th

e su

rger

y

Chapter 11

Positive end – expiratory pressure following coronary artery bypass grafting

Dongelmans DA, Hemmes SNT, Kudoga AC, Veelo DP, Binnekade JM, Schultz MJMinerva Anestesiologica 2012 ;78(7):790-800

252

Abstract

Background. Cardiac surgery–related pulmonary complications include alterations in lung mechanics and anomalies in gas exchange. Higher levels of positive end–expiratory pressure (PEEP) have been suggested to benefit cardiac surgical patients. We compared respiratory compliance, arterial oxygenation and time till tracheal extubation in 2 cohorts of patients weaned from mechanical ventilation with different levels of PEEP after elective and uncomplicated coronary artery bypass grafting (CABG). We hypothesized that higher PEEP levels improve pulmonary compliance and gas exchange in the first hours of weaning from mechanical ventilation, but not to shorten time till tracheal extubation.

Materials and Methods. Secondary retrospective analysis of 2 randomized controlled trials: in the first trial patients were weaned with PEEP levels of 10 cmH2O for the first 4 hours followed by PEEP levels of 5 cmH2O until tracheal extubation (high PEEP, HP); and the second trial patients were weaned with PEEP levels of 5 cmH2O during the entire weaning phase (low PEEP, LP). The primary endpoint was pulmonary compliance. Secondary endpoints included arterial oxygenation, duration of mechanical ventilation and post-operative pulmonary complications.

Results. The analysis included 121 patients; 60 HP patients and 61 LP patients. Baseline characteristics were similar. Compared to LP patients, HP patients had a better pulmonary compliance, 47.2 ± 14.1 versus 42.7 ± 10.2 ml/cmH2O (P < 0.05), and higher levels of PaO2, 18.5 ± 6.6 (138.75 ± 49.5) versus 16.7 ± 5.4 (125.25 ± 40.5) kPa (mmHg) (P < 0.05). Patients in the HP group were less frequent in need of supplementary oxygen after ICU discharge. These differences remained present during the entire weaning phase, even after reduction of PEEP. However, HP patients had a longer time till tracheal extubation, 16.9 ± 6.1 versus 10.5 ± 5.0 hours (P < 0.001). HP patients had longer durations of postoperative infusion of propofol, 4.9 [2.6 – 7.4] versus 3.5 [1.8 – 5.8] hours (P < 0.05). There were no differences in use of inotropes. Cumulative fluid balances were slightly higher in HP patients.

Conclusion. Use of higher PEEP levels after elective uncomplicated CABG improves pulmonary compliance and oxygenation but seems to be associated with a delay in tracheal extubation.

PEEP following CABG

Chap

ter

11

253

Introduction

Pulmonary dysfunction is a ubiquitous consequence of cardiac surgery.1 Cardiac surgical patients are subjected to surgery–related factors, including sternotomy, cooling, the use of cardiopulmonary bypass and internal mammary artery dissection, which all predispose them to postoperative pulmonary complications. Cardiac surgery–related pulmonary complications include alterations in lung mechanics, anomalies in gas exchange, or both.1 Alterations in the mechanical properties of the lung lead to reductions in pulmonary compliance,2 vital capacity3 and functional residual capacity,4 which are reflected by immediate but short–lasting changes of the inspiratory pressure–volume curve of the respiratory system.5 Indeed, up to 4 hours after cardiac surgery the inspiratory pressure–volume curve has a sigmoid shape because of a right–shift of the lower inflection point (LIP) and a down–shift of the upper inflection point.

A positive end–expiratory pressure (PEEP) level set above the LIP may prevent atelectasis and decrease small airway closure.6,7 As such, higher PEEP levels could benefit cardiac surgical patients. However, higher PEEP levels could also have negative effects. Higher PEEP levels may cause caregivers to give more sedatives, which could lengthen the weaning process. In addition, higher PEEP levels may compromise cardiac output,8 mandating the use of more intravenous fluids to increase the afterload and inotropes to improve cardiac performance.

The aim of this study was to scrutinize the effects of higher PEEP levels on pulmonary function and weaning of cardiac surgical patients. We hypothesized that higher PEEP levels improve pulmonary compliance and gas exchange in the first hours of weaning from mechanical ventilation, but not to shorten time till tracheal extubation. Hereto we reanalysed and compared data from 2 randomized controlled trials of patients after elective and uncomplicated coronary artery bypass grafting (CABG) in which different PEEP levels were used.9,10

Methods

Study designIn this retrospective analysis, we analysed and compared prospectively collected respiratory parameters; times till tracheal extubation, prescriptions of sedatives and inotropes, and cumulative fluid balances from 2 randomized controlled trials of patients after CABG.9,10 The first trial was performed from October 2005 to July 2006,9 the second trial from August 2007 to August 2008.10 The mechanical ventilation protocol in the intervention arm of the first trial9 only differed from that in the control arm of the second trial10 with respect to the PEEP level used in the first 4 hours of weaning from mechanical ventilation (see below). In addition, we collected and compared need for supplementary oxygen and peripheral oxygen saturations in the ward, the presence or absence of infiltrates, atelectasis, pleural effusion and signs of fluid overload on chest X–rays, and the presence or absence of pneumonia.

SettingBoth trials recruited patients after CABG admitted to the 28–bed intensive care unit (ICU) of

254

the Academic Medical Center, Amsterdam, the Netherlands. From October 2005 until August 2008 neither the medical staff nor the nursing staff changed. The medical staffing consisted of 8 full–time intensivists, 8 ICU fellows and 10 residents of other specialties, such as internal medicine, anaesthesiology and surgery. The nursing staff consisted of 140 qualified ICU nurses.

The protocols for anesthesia during surgery and post–operative care in the ICU were also left unaltered during conduct of both trials. In short, anesthesia started with 1 or 2 mg lorazepam as pre–medication, which was followed by etomidate, sufentanil and rocuronium for induction of anesthesia and facilitation of intubation. During the surgical procedure small doses of sufentanil were used as analgesic, and sevoflurane plus propofol were used to maintain anesthesia. Muscle relaxants were not given during the surgical procedure. Small boluses of morphine and midazolam could be given at the end of the procedure. Cardiopulmonary bypass was performed under moderate hypothermia (28 – 35°C), using a membrane oxygenator and non–pulsatile blood flow. At the end of anesthesia, all patients were transferred to the ICU with tracheal intubation. In the ICU fluid resuscitation existed of intravenous infusion of normal saline and starch solutions, and blood transfusion to maintain hemoglobin concentration (≥ 5.0 mmol/L or 190 g/l). Dopamine and norepinephrine were infused to achieve mean arterial pressure ≥ 70 mmHg, dobutamine and/or enoximone were infused to achieve a cardiac index ≥ 2.5 L/min/m2 or a mixed venous oxygenation > 60%. Propofol was given for sedation via continuous infusion until core temperature was > 36.00C. Acetaminophen (4 gram/day) was started in all patients. Morphine was given in small boluses of 1 to 2 mg intravenously at the descretion of attending ICU nurses. No neuromuscular blocking agents were used in the ICU.

The original trialsThe local institutional review board approved both trials, and preoperative written and signed informed consent was obtained from eligible patients programmed for elective CABG.

Inclusion and exclusion criteria were similar for both trials. In both trials we created a homogenous group of patients of ≥ 18 years after elective and uncomplicated CABG. Patients with a history of pulmonary disease or a history of pulmonary surgery were excluded; patients with an intra–aortic balloon pump or inotropes and/or vasopressors at a more then usual rate (in mg per hour): dopamine (16), norepinephrine (4), dobutamine (20) or epinephrine [any rate]) on arrival in the ICU were also excluded.

All patients were ventilated by Hamilton Galileo ventilators in the adaptive support ventilation (ASV) mode.11 (software version GMP03.41f, GCP03.40a, GTP01.00; Hamilton Medical AG, Rhäzüns, Switzerland). There were no differences in software versions between the original trails. Passive humidification of the ventilatory circuit was applied by means of a HME–filter (Medisize Hygrovent S, Medisize, Hillegom, The Netherlands). Minute ventilation was set at 100% of the theoretical value based on ideal body weight (IBW) (“100% minute ventilation”) and oxygen inspiratory fraction (FiO2) of 50%. Maximum airway pressure (P–max) was set 35 cmH2O (high–pressure pop–off). Flow trigger sensitivity was set at 2 L/sec. An arterial blood gas (ABG) analysis (Rapid Lab 865; Bayer Diagnostics, Dublin, Ireland) was performed 30 minutes after connection to the ventilator. If PaCO2 was < 3.5 kPa (26.25 mmHg) or > 5.5 kPa (41.25 mmHg) minute ventilation was adjusted. Any modification of the ventilator settings was advised to be

PEEP following CABG

Chap

ter

11

255

controlled after 30 minutes by a new ABG analysis. FiO2 was adjusted to maintain arterial oxygen saturation (SaO2) of ≥ 95%. The patient was extubated after achieving general tracheal extubation criteria (i.e., responsive and cooperative, urine output > 0.5 ml/kg/h, chest tube drainage < 100 ml last hour, no uncontrolled arrhythmia, rectal temperature > 360C, spontaneous respiratory frequency of 10–20 breaths per minute, futhermore a FiO2 of 40% and an inspiratory pressure (P–insp) of 5 to 10 cmH2O for 30 minutes.

PEEP level settingsIn the intervention group of the first trial,9 the PEEP level was set at 10 cmH2O in the first 4 hours after arival in the ICU, and therafter at 5 cmH2O until tracheal extubation (high PEEP, HP). In the control group of the second trial,10 the PEEP level was set at 5 cmH2O from admission to the ICU until tracheal extubation (low PEEP, LP).

Escape ventilation modeIn both trials pressure controlled and pressure support ventilation were indicated as escape ventilation if patients would fail weaning with ASV or afer reintubation.

Data collectionThe following data were collected: demographic data (gender, age, height, weight) operation characteristics (duration of anesthesia, duration of cardiopulmonary bypass, duration of cross–clamping).

Ventilation parameters (PEEP level, respiratory rate and tidal volume (VT), P–insp and dynamic compliance [all collected by a data logger (Hamilton data logger, version 3.27.1, Hamilton Medical AG connected to the ventilator], blood gas analysis results).

Outcome data (time till tracheal extubation, re–intubation, and length of stay in ICU [collected from the Patient Data Management System, IMDsoft, Sassenheim, the Netherlands]).

Other postoperative data: need of oxygen in the ward, rate of atelectasis at chest X-ray and pneumonia.

ICU data. Hourly fluid balances in three study fases, use of inotropics, opiod dose and types, midazolam dose, propofol dose. ICU discomfort data (use of physical restraints, the occurences of stress and/or agitation, as reported in medical and/or nursing reports).

Time phasesFor comparisons between groups, we defined 3 phases during stay in ICU. In HP patients, “phase–1” was the period during which patients were ventilated with a PEEP level of 10 cmH2O; “phase–2” was the period during which patients were ventilated with a PEEP level of 5 cmH2O (i.e., untill tracheal extubation); “phase–3” was the period from tracheal extubation untill discharge from the ICU. Phase–1 for LP patients was defined as the mean time of “phase–1” in HP patients; “phase–2” was the following period that ended with tracheal extubation; “phase–3” was the period from tracheal extubation untill discharge from the ICU.

256

Table 1. Patient characteristics

Variable HP patients N = 63

LP patients N = 63

Gender, male (%) 55 (87) 55 (87)

Age, years 65 ± 8 64 ± 9

Actual body weight, kg 85 ± 14 83 ± 10

Predicted body weight, kg 71 ± 9 71 ± 8

Number of bypasses, N 3 [3 – 4] 3 [3 – 4]

Use of arterial graft, N (%) 51 (81%) 44 (70%)

Pump time, minutes 97 ± 33 103 ± 36

Aortic clamp time, minutes 59 ± 25 63 ± 27

Opiates OR – total dose, mg/kg 2.5 [1.8 – 3.4] 2.5 [2.0 – 3.1]

Benzodiazepines OR – total dose, mg/kg 1.7 [0.4 – 2.5] 1.8 [0.5 -2.8]

Length of stay in ICU, hours 25 [22 – 63] 27 [22 – 40]

HP: high positive end–expiratory pressure (PEEP); LP: low PEEP; ICU: intensive care unit; OR: Operation room. Data are means ± SD or medians [IQR]

Table 2. Mechanical ventilation characteristics

Variable Phase HP patientsN = 60

LP patientsN = 61 p value

Respiratory rate, breath/min1 13.9 ± 1.8 13.8 ± 2.0 0.855

2 12.9 ± 1.4 14.5 ± 2.3 < 0.05

Tidal volume, ml1 613 ± 98 582 ± 92 < 0.05

2 579 ± 105 589 ± 100 0.10

Tidal volume, ml/kg IBW1 8.6 ± 0.9 7.1 ± 1.1 < 0.05

2 8.4 ± 1.3 8.4 ± 1.2 0.87

P–insp, cmH2O1 12.7 ± 3.1 13.6 ± 2.8 0.28

2 11.9 ± 3.1 13.2 ± 3.1 0.45

Compliance ml/cmH2O 1 47.2 ± 14. 1 42.7 ± 10. 2 < 0.05

2 48.6 ± 13. 2 44.6 ± 11. 3 0.33

FiO2 1 42.2 ± 3 42.4 ± 5 0.75

2 42.6 ± 3 41.6 ± 7 0.27

HP: high positive end–expiratory pressure (PEEP); LP: low PEEP; IBW, ideal body weight; P–insp: inspiratory pressure; FiO2: fraction of inspiration oxygen. Data are means ± SD or medians [IQR]

PEEP following CABG

Chap

ter

11

257

DefinitionsIdeal body body weight (IBW) was calculated by the following formula: in men, IBW (kg) = 50 + 0.91 X (centimeters of height - 152.4); in women, IBW (kg) = 45.5 + 0.91 X (centimeters of height - 152.4).12

Hourly cummulative fluid balances were calculated, ignoring insensible loss. Cumulative fluid balance was defined as the sum of all fluids (fluids in and out the patient) this was calculated per time-phase (see above). Inotrope doses were compared using a previously described inotrope score.13 The inotrope score was calculated as dopamine (x1) + dobutamine (x1) + milrinone (x15) + norepinephrine (x100).

Opiate doses were all recalculated as morphine equipotent doses with the following formula: 10 miligram (mg) of morphine = 0.1 mg fentanyl = 0.01 mg sufentanil.14 Doses of benzodiazepines were similary converted to equipotent doses of diazepam using the following formula: 5 mg midazolam = 10 mg diazepam = 50 mg oxazepam.15 Patient discomfort was defined as stress and agitation reported in the patient’s medical chart that led to an intervention such as administration of a benzodiazepine or mobilization.

We defined postoperative pneumonia as: new infiltrate(s) on the chest x-ray with leucocytes above 12 x109, fever (temperature above 38.3) and purulent sputum.

Statistical analysisDescriptive statistics were used to summarize patient characteristics. Categorical variables were compared between groups by chi–square tests. If normally distributed, continuous values are expressed as means ± standard deviation (SD), otherwise medians and interquartile ranges [IQR] were used. All analyses were performed in SPSS version 16.0 (SPSS inc., Chicago, IL).

Results

PatientsIn the original trials 64 patients were randomized to the intervention arm of the first trial9 and 64 patients to the control arm of the second trial.10 In both arms 1 patient was lost to analysis due to reaching exclusion criteria with respect to use of inotropes. Three HP patients and 2 LP patients were lost to analysis due to datalogger failure. This left us with 60 HP patients and 61 patients LP patients. Patient characteristics are shown in table 1. Patients from the 2 trials were well matched according to their baseline characteristics. No significant differences were found in baseline characteristics as well as in intra–operative medication requirements (aesthetics, opioids and benzodiazepines). Body temperature on arrival in the ICU was similar.

The PEEP level was set at 10 cmH2O for 5.0 ± 2.5 hours in HP patients, thereafter the PEEP level was maintained at 5 cmH2O until tracheal extubation. In LP patients PEEP levels were never set above 5 cmH2O.

258

Table 3. Prescribed sedatives

Variable Phase HP patientsN = 60

LP patientsN = 61 p value

Total propofol dose, mg/kg

1 5.9 [3.0 – 9.7] 2.2 [5.2 – 8.0] 0.427

2 0 [0 – 4.0] 0 [0 – 2.1] 0.162

3 0 0 1

Propofol treatment, hours

1 4.9 [2.6 – 5.0] 3.5 [0.94 – 1.6] < 0.05

2 0 [0 – 2.5] 0 [0 – 1.0] 0.110

3 0 0 1

Total morfine dose, mg

1 0 [0 – 0] 0 [0 – 2.5] 0.390

2 0 [0 – 2.4] 0 [0 – 0] 0.199

3 0 [0 – 0] 0 [0 – 2.9] 0.064

Total benzodiazepines dose, mg

1 0 [0 – 0] 0 [0 – 0] 0.321

2 0 [0 – 0] 0 [0 – 0] 0.151

3 0 [0 – 0] 0 [0 – 0] 0.353

HP: high positive end–expiratory pressure (PEEP); LP: low PEEP. Data are means ± SD or medians [IQR]

Table 4. Discomfort data

Variable HP patientsN = 60

LP patientsN = 61 p value

Use of physical restraints, number of patients 9 3 0.06

Stress and/or agitation, number of patients 9 13 0.39

HP: high positive end–expiratory pressure (PEEP); LP: low PEEP. Data are means ± SD or medians [IQR]

Table 5. Inotrope scores and cumulative fluid balances

Variable Phase HP patientsN = 60

LP patientsN = 61 p value

Inotrope score

1 101 [15 – 115] 100 [5 – 115] 0.529

2 100 [100 – 115] 15 [0 – 115] 0.956

3 0 [0 – 0] 0 [0 – 0] 0.365

Cumulative fluid balance, ml

1 1112 ± 1108 1130 ± 999 0.928

2 776 ± 1127 507 ± 1054 0.198

3 643 ± 834 177 ± 738 < 0.05

HP: high positive end–expiratory pressure (PEEP); LP: low PEEP. Data are means ± SD or medians [IQR]

PEEP following CABG

Chap

ter

11

259

While there were no HP patients who needed reintubation, 2 LP patients were reintubated after planned extubation (p = 0.87).

Both patients had to be reintubated, probably due to atelectasis both confirmed on chest x-ray after reintubation and ventilated with the escape ventilation modes pressure controlled and pressure support. Both patients were extubated in the subsequent course of their ICU stay.

Pulmonary complianceMechanical ventilation characteristics are presented in table 2. Compliance was higher in the HP patients in phase–1; although compliance remained higher in HP patients in phase–2, the difference did no longer reach statistical significance. VT was larger in HP patients in phase–1 and 2; P–insp was lower in phase–1 and 2, although differences did not reach statistical significance.

Arterial oxygenation and carbon dioxide levelsBlood gas analysis data are shown in figure 1. PaO2 levels were higher in HP patients in phase–1 and phase–2, but not in phase–3. In accordance, PaO2 to FiO2 ratios were higher in HP patients in phase–1, and remained significantly higher in phase–2. PaCO2 levels were lower in HP patients

Figure 1. Arterial oxygenation (PaO2), arterial carbon dioxide (PaCO2) and arterial pH, in phase–1, 2 and 3, and PaO2 to FiO2 ratios (P/F) in phase–1 and 2 after admission to ICUClosed symbols, patients ventilated with higher PEEP levels in phase–1; open symbols, patients ventilated with lower PEEP levels during the entire weaning period. *denotes statistical significant differences between groups

260

in phase–1 and phase–2. Arterial pH was similar between the 2 study groups, in all predefined phases. Patients in the HP group were less frequently in need for supplementary oxygen in the ward (relative risk of 0.80 (0.66 – 0.96), p-value of 0.02). Peripheral oxygen saturations in the ward were not different. There were no differences between groups regarding atelectasis, pleural effusion and signs of fluid overload at chest X–rays on day 1, two or three postoperatively. Two patients in the HP group developed pneumonia.

Duration of mechanical ventilationDuration of mechanical ventilation is shown in figure 2. Time from ICU admission to tracheal extubation was different between groups. HP patients were extubated after 16.9 ± 6.1 hours, while LP patients were extubated after 10.5 ± 5.0 hours (p < 0.001).

Sedatives and patient discomfortDose and duration of sedatives are shown in table 3. Discomfort data are shown in table 4. Median cumulative dose of propofol administered during stay in ICU tended to be higher in the HP patients, 8.5 [3.9 – 14.9] versus 5.8 [2.6 – 11.8] mg/kg (p = 0.16). In addition, propofol infusion was continued for a longer time, 4.9 [2.6 – 7.4] versus 3.5 [1.8 – 5.8] hours (p < 0.05). In phase–1 there was a significant difference between groups 4.9 [2.6 – 5.0] hours in the HP patients versus 1.6 [0.94 – 3.5] hours in the LP patients (p < 0.05). Opiates and benzodiazepines were seldom administered in both groups. Discomfort was not different between HP and LP patients.

Figure 2. Kaplan–Meier curve showing time until tracheal extubationClosed line, patients ventilated with higher PEEP levels in phase–1; Dotted line, patients ventilated with lower PEEP levels during the entire weaning period

PEEP following CABG

Chap

ter

11

261

Table 6. Postoperative data

Variable HPN=60

LPN=61 RR (95% CI) p value

Sputum 1 3 3 (0.32 / 28.05) 0.62

Fever 9 9 1 (0.43 / 2.35) 1.0

Leucocytes 38 36 0.95 ( 0.71 / 1.26) 0.85

O2 54 43 0.80 (0.66 / 0.96) < 0.05

Antibiotics_pulm 0 0 - -

Pneumonia 0 2 - -

Day 1 (n) CXR 6 (9) 7 (9) 1.17 (0.65 / 2.08) 1.0

Day 2 (n) CXR 41(47) 29 (25) 0.99 (0.82 / 1.19 ) 1.0

Day 3 (n) CXR 33 (36) 32 (37) 0.94 (0.80 / 1.11) 0.71

Day 1 (n) Atelectasis 4 (9) 6(9) 1.50 (0.63 / 3.56) 0.63

Day 2 (n) Atelectasis 31 (47) 20 (29) 1.05 (0.76 / 1.44) 0.98

Day 3 (n) Atelectasis 24 (35) 28 (36) 1.10 (0.83 / 1.47) 0.62

Day 1 (n) Saturation 96 (42) 96 (29) - 0.36

Day 2 (n) Saturation 94 (56) 94 (43) - 0.59

Day 3 (n) Saturation 94 (58) 94 (46) - 0.59

HP: high positive end–expiratory pressure (PEEP); LP: low PEEP; N: number of patients; O2: Oxygen need on the ward; Antibiotics_pulm: Antibiotics prescribed for suspected pneumonia; n: number of occurrence; CXR: Chest X-ray abnormalities; Atelectasis on chest X-ray; Saturation: peripheral oxygen saturation in percentage

Inotropes and cumulative fluid balancesPrescription of inotropes and cumulative fluid balances are shown in table 5. The inotrope score was higher in HP patients in phase–2, but differences did not reach statistical significance. There were no clinically important differences between the 2 study groups with respect to cumulative fluid balances, although HP patients had a significant higher cumulative fluid balance at the end of stay in ICU.

Postoperative complicationsNeed of oxygen in the ward was significantly different between groups. Patients in the HP group were less frequent in need of supplementary oxygen in the normal ward, after ICU discharge (relative risk of 0.80 (0.66 – 0.96), p = 0.02). There were however no differences between groups regarding atelectasis at chest X-ray and other postoperative data (table 6). There were, according to our definition, two cases of pneumonia the HP group.

262

Discussion

In this secondary analysis of 2 randomized controlled trials of patients after elective and uncomplicated CABG we determined the effects of use of higher PEEP levels on mechanical ventilation characteristics, including pulmonary compliance and oxygenation. The results can be summarized as follows: (1) compliance was higher in HP patients in phase–1, but not in phase–2; (2) oxygenation was better in HP patients in phase–1 and 2, but similar in phase–3; (3) time till tracheal extubation was longer in the HP patients and (4) Patients in the HP group needed supplementary oxygen after ICU discharge less frequently.

Our analysis confirms results from previous studies suggesting that higher PEEP levels improve pulmonary function after cardiac surgery.5–7 Indeed, with the use of higher PEEP levels pulmonary compliance was better and arterial oxygenation improved. Notably, the beneficial effect on pulmonary compliance was only found in phase–1, and the effect on arterial oxygenation disappeared after tracheal extubation. This latter finding is in accordance with findings of a previous observational study and randomized controlled trials.5,16,17 In one observational study of cardiac surgical patients, changes in lung mechanics were only present in the first hours after surgery.5 In a randomized controlled trail of patients after CABG, in which patients were mechanical ventilated with PEEP levels of 0, 5 or 10 cmH2O, it was shown that use of higher PEEP levels offered no sustained beneficial effect on arterial oxygenation or the occurrence of atelectasis on chest X–rays.16 In another randomized controlled trial of cardiac surgical patients, in which patients were mechanically ventilated with PEEP levels titrated on the best achievable PaO2 level, a similar course of the effects of PEEP over time was noted.17 It should be noted that patients in the HP group needed less frequently supplementary oxygen after ICU discharge. This may be because of prevention of atelectasis. Atelectasis may have been too small to be detected by chest X-rays, explaining why we did not find a difference between the study groups with regard to atelectasis on chest X–rays. Higher PEEP levels are frequently used in mechanically ventilated patients suffering from acute lung injury. Two recent metaanalyses of randomized controlled trials of patients with acute lung injury showed higher PEEP to be associated with a reduction in hospital mortality.18,19 Notably, this effect was only found in patients with more severe acute lung injury, since patients with less severe acute lung injury had no clinical benefit from the use of higher PEEP levels. Thus, the beneficial effects of PEEP seem to depend on the severity of illness. Obviously, short-lived pulmonary dysfunction, as found in patients after cardiac surgery, is different from abnormalities found in patients with acute lung injury.

We did not expect the PEEP level to have an effect on time till tracheal extubation after elective uncomplicated CABG. We found nevertheless a significant difference in time till extubation. Differences in the use of sedatives could explain differences in time to extubation. Sedation is often needed during the time the patients spend on the ventilator and are warming up, however oversedation can prolong this period and lead to adverse effects for the patient.20 We found that although the total dose of propofol was the same, the duration of the use of propofol was longer in the HP patients as well overall as in phase–1. This suggests that the use of higher PEEP leads to longer use of sedatives and this, in turn, could contribute to a longer duration of ventilation. However since the difference in time till extubation is much larger (almost 6 hours) then the difference in the time of the use of propofol (almost 1.5 hours) it is unlikely that the

PEEP following CABG

Chap

ter

11

263

longer duration of the use of propofol explains the total difference in duration of ventilation. In a post hoc regression analysis we calculated that the attribution to the time till extubation of the duration of propofol infusion was 18% in the HP group and 8% in the LP group.

Another reason for the differences in duration of ventilation could be differences in the use of inotropes. However we found no differences in inotrope scores between HP and LP patients suggesting no influence of the applied PEEP level on the use of inotropes. Moreover it should be noted that although the protocol stated that HP should be given during 4 hours the mean duration of HP was 5 ± 2,5 h. The reason for this could be that PEEP was to be lowered according to the guideline when the patient was stable. Determining this could include a new blood gas analysis thus leading to delay.

As a high PEEP level may affect venous return, we expected a more positive fluid balance to maintain preload of the heart in the high PEEP group. Indeed, the ICU–guideline clearly advised to first infuse fluids before increasing the infusion rate of inotropics. A difference in fluid balances from ICU admission till tracheal extubation was not found. In both groups the fluid balance from ICU admission till tracheal extubation was approximately one liter, which is expected in patients who arrive in the ICU with mild hypothermia and sedation. The difference in fluid balance after tracheal extubation remains unexplained. Notably, the this difference was not associated with differences in oxygenation. In the postextubation period on the ward there were significant differences in the number of patients needing oxygen therapy. The LP group needed more oxygen. Whether or not this was caused by more atelectasis in this group as one would suspect is difficult to asses. The number of patients with atelectasis on there chest X-ray did not differ between the groups.

Although 2 patients in the HP group developed pneumonia, while no pneumonia was observed in the LP group, it should be noted that this difference was not statistically significant. If we added use of antibiotics for any cause (or the clinical diagnosis of pneumonia) to our definition no cases of pneumonia were present in both groups.

There are several limitations to our analysis. It should be recognized that the design of our study is not the most appropriate for comparison of two treatment strategies. This is an important drawback of our study, which should be taken into account when interpreting the study results. It would have been better to perform a randomized clinical trial to minimize the chances on bias and confounders.

Our analysis should be seen as a before–after study. It might be that our practice changed over time but that this change remained unnoted. However it should be said that the OR and ICU staff remained largely similar. Also, aside from the PEEP level used in mechanically ventilated patients after cardiac surgery, no changes were introduced in the local mechanical ventilation guideline. Moreover the question should be raised whether tracheal extubation of cardiac surgical patients is dependent on the ventilatory strategy e.g. PEEP levels alone or (also) on factors independent on the ventilation strategy. Anaesthesia and ICU teams may gain experience in the treatment of these patients, which could hasten tracheal extubation.21 Also, increased experience with the ventilation mode used in the two studies (Adaptive Support Ventilation) could have altered

264

the treatment of these patients. It should be noted, however, that our team started the use of Adaptive Support Ventilation in 2004. And finally, better awareness of rather long weaning times in our institution could have led to a more proactive behaviour with regard to tracheal extubation.22

Another limitation is the titration of PEEP in our study. Notably, the PEEP level in our patients was not titrated on the LIP of individual pressure volume curves, since interpretation of pressure volume curves can be difficult and time consuming.23-27 We rather choose for a simple approach to use a PEEP level of 10 cmH2O for 4 hours, based on the findings in a previous observational study.5 This practical approach could have led to the use of too low PEEP levels. This in turn could have led to insufficient effect of the high PEEP strategy while in the meantime resulting in longer weaning times. Contrary our approach could also have led to unnecessary high levels of PEEP. Resulting in an exaggeration of the possible negative effects of higher PEEP levels and again to longer weaning times. Either way thus resulting in longer weaning times without the benefits of the use of higher PEEP levels.

While none of the HP patients needed to be reintubated, 2 LP patients had to be reintubated. Although this difference was not statistically significant, the finding is though–provoking. From the present analysis it cannot be concluded that the higher reintubation rate in LP patients is causally related to the PEEP level used in the first hours of weaning. However, since the 2 reintubated patients needed continuation of mechanical ventilation because of atelectasis, it can at least be speculated that the higher PEEP levels could prevent reintubation. It is legitimate to ask ourselves what is preferable: To extend few hours the period of weaning and prevent extubation failure or to shorten the routine weaning process and increase the risk of extubation failure? The fact that more LP patients used oxygen in the ward fits this picture as well. This hypothesis needs however to be tested in a well–powered randomized controlled trial

Conclusion

Use of higher PEEP levels after elective uncomplicated CABG improves pulmonary compliance and oxygenation but is associated with a delay in tracheal extubation.

PEEP following CABG

Chap

ter

11

265

References1. Wynne R, Botti M. Postoperative pulmonary dysfunction in adults after cardiac surgery with cardiopulmonary bypass:

clinical significance and implications for practice. Am J Crit Care 2004; 13(5): 384-932. Chaney MA, Nikolov MP, Blakeman B, Bakhos M, Slogoff S. Pulmonary effects of methylprednisolone in patients

undergoing coronary artery bypass grafting and early tracheal extubation. Anesth Analg 1998; 87(1): 27-333. Johnson D, Hurst T, Thomson D, Mycyk T, Burbridge B, To T, Mayers I. Respiratory function after cardiac surgery. J

Cardiothorac Vasc Anesth 1996; 10(5): 571-74. Shapira N, Zabatino SM, Ahmed S, Murphy DM, Sullivan D, Lemole GM. Determinants of pulmonary function in patients

undergoing coronary bypass operations. Ann Thorac Surg 1990; 50(2): 268-735. Ranieri VM, Vitale N, Grasso S, Puntillo F, Mascia L, Paparella D et al. Time-course of impairment of respiratory

mechanics after cardiac surgery and cardiopulmonary bypass. Crit Care Med 1999; 27: 1454-606. McCarthy GS, Hedenstierna G. Arterial oxygenation during artificial ventilation. The effect of airway closure and of its

prevention by positive end-expiratory pressure. Acta Anaesthesiol Scand 1978; 22: 563-97. West JB. First annual SCCM lecture: pulmonary gas exchange in the critically ill patient. Crit Care Med 1974; 2: 171-808. Luecke T, Pelosi P. Clinical review: Positive end-expiratory pressure and cardiac output. Crit Care 2005; 9: 607-219. Dongelmans DA, Veelo DP, Paulus F, de Mol BA, Korevaar JC, Kudoga A et al. Weaning automation with adaptive support

ventilation: a randomized controlled trial in cardiothoracic surgery patients. Anesth Analg 2009; 108: 565-7110. Dongelmans DA, Veelo DP, Binnekade JM, de Mol BA, Kudoga A, Paulus F, Schultz MJ. Adaptive support ventilation

with protocolized de-escalation and escalation does not accelerate tracheal extubation of patients after nonfast-track cardiothoracic surgery. Anesth Analg 2010; 111(4): 961-7

11. Brunner JX. Iotti GA. Adaptive Support Ventilation (ASV). Minerva Anestesiologica 2002; 68(5): 365-36812. The Acute Respiratory Distress Syndrome Network.Ventilation with lower tidal volumes as compared with traditional

tidal volumes for acute lung injury and the acute respiratory distress syndrome. NEJM 2000; 342(18): 1301-813. Wernovsky G, Wypij D, Jonas RA, Mayer JE Jr, Hanley FL, Hickey PR et al. Postoperative course and hemodynamic

profile after the arterial switch operation in neonates and infants. A comparison of low-flow cardiopulmonary bypass and circulatory arrest. Circulation 1995; 92: 2226-35

14. Reisine PG. The Pharmacological Basis of Therapeutics. Ninth Edition. ed. New York: McGraw-Hill, 199615. Wilson WC, Smedira NG, Fink C, McDowell JA, Luce JM et al. Ordering and administration of sedatives and analgesics

during the withholding and withdrawal of life support from critically ill patients. JAMA 1992; 267: 949-5316. Marvel SL, Elliott CG, Tocino I, Greenway LW, Metcalf SM, Chapman RH. Positive end-expiratory pressure following

coronary artery bypass grafting. Chest 1986; 90(4): 537-4117. Celebi S, Köner O, Menda F, Omay O, Günay I, Suzer K et al. The pulmonary and hemodynamic effects of two different

recruitment maneuvers after cardiac surgery. Anesth Analg 2007; 104: 384-9018. Briel M, Meade M, Mercat A, Brower RG, Talmor D, Walter SD, Slutsky AS et al. Higher vs lower positive end-expiratory

pressure in patients with acute lung injury and acute respiratory distress syndrome: systematic review and metaanalysis. JAMA 2010; 303(9): 865-73

19. Putensen C, Theuerkauf N, Zinserling J, Wrigge H, Pelosi P. Metaanalysis: ventilation strategies and outcomes of the acute respiratory distress syndrome and acute lung injury. Ann Intern Med 2009; 151(8): 566-76

20. Kollef MH, Levy NT, Ahrens TS, Schaiff R, Prentice D, Sherman G. The use of continuous i.v. sedation is associated with prolongation of mechanical ventilation. Chest 1998; 114: 541-8

21. De Hert SG, Van der Linden PJ, Cromheecke S, Meeus R, ten Broecke PW, De Blier IG, Stockman BA, Rodrigus IE. Choice of primary anesthetic regimen can influence intensive care unit length of stay after coronary surgery with cardiopulmonary bypass. Anesthesiology 2004; 101(1): 9-20

22. Hawkes CA, Dhileepan S, Foxcroft D, Imberger G. Early extubation for adult cardiac surgical patients. Cochrane Database of Systematic Reviews 2003, Issue 4. Art. No.: CD003587

23. Pestana D, Hernández-Gancedo C, Royo C, Una R, Villagrán MJ, Pena N, Criado A. Adjusting positive end-expiratory pressure and tidal volume in acute respiratory distress syndrome according to the pressure-volume curve. Acta Anaesthesiol Scand 2003; 47(3): 326-34

24. Jonson J, Svatensson C. Elastic pressure–Volume curves: what information do they convey? Thorax 1999; 54: 82–725. Lichtwark-Aschoff M, Mols G, Hedlund AJ, Kessler V, Markström AM, Guttmann J et al. Compliance is nonlinear over

tidal Volume irrespective of positive end-expiratory pressure level in surfactant-depleted piglets. Am J Respir Crit Care Med 2000; 162: 2125–33

26. Jonson B, Richard J-C, Straus C, Mancebo J, Lemaire F, Brochard L. Pressure-Volume curves and compliance in acute lung injury. Evidence of recruitment above the lower inflection point. Am J Respir Crit Care Med 1999; 159: 1172–78

27. Hickling KG. Best compliance during a decremental, but not incremental, positive end-expiratory pressure trial is related to open lung positive end-expiratory pressure. A mathematical model of acute respiratory distress syndrome lungs. Am J Respir Crit Care Med 2001; 163: 69–78

Chapter 12

Summary and general discussion

Hemmes SNT

268

Introduction

As an introduction to this chapter, we briefly summarize findings published before the writing of the thesis titled: ‘lung-protective perioperative mechanical ventilation’. A more extensive overview of this literature is provided in chapter 1.

Postoperative pulmonary complicationsSimilar to mechanical ventilation in critically ill patients, intraoperative ventilation has a strong potential to cause lung injury.1 Certain intraoperative ventilation settings are associated with the development of pulmonary complications after surgery.2 Occurrence of so–called postoperative pulmonary complications is strongly associated with clinical outcome.3

Ventilation–associated lung injuryIt is generally believed that overdistension and repetitive opening and closing of lung tissue are the two main mechanical causes of so–called ‘ventilation–associated lung injury’ (VALI).4 Use of too large tidal volumes could cause overdistension of lung tissue that remains aerated at the end of expiration, which could be aggravated by too high levels of positive end–expiratory pressure (PEEP). Too low levels of PEEP, though, could result in repetitive opening and closing of lung tissue that collapses at the end of expiration. Next to ‘mechanical stress’, ventilation could also cause ‘chemical stress’, as too high levels of fractional inspired oxygen (FiO2) can increase the production of reactive oxygen species (ROS),5 which have a direct toxic effect on lung cells. Too high levels of FiO2 could also increase the risk of resorption atelectasis.6 Lung-protective mechanical ventilation aims at low tidal volume ventilation to prevent overdistention, and higher levels of PEEP with recruitment manoeuvres to prevent repetitive collapse. Additionally, restrictive levels of FiO2 could protect against hyperoxia-induced lung injury.

Prevention of lung injury in critically ill patientsTwo large metaanalyses convincingly confirmed the results from randomized controlled trials in critically ill patients with the acute respiratory distress syndrome (ARDS) that showed ventilation with low tidal volumes to improve morbidity and mortality.7,8 While three randomized controlled trials in patients with ARDS individually showed no benefit of higher levels of PEEP,9-11 one individual patient data metaanalyses strongly suggest higher levels of PEEP to improve outcome of patients with moderate or severe ARDS.7

The evidence for benefit of using low tidal volumes in critical care patients without ARDS is less convincing, with only one randomized controlled trial in these patients showing a lower incidence of VALI during low tidal volume ventilation.12 One conventional metaanalysis13 and two individual patient data metaanalyses14,15 confirm these findings, but the quality of the included studies was sometimes low, hampering firm conclusions. Whether patients without ARDS benefit from higher levels of PEEP, is even more uncertain, with one trial suggesting benefit,16 one trial showing no effect,17 and one trial even suggesting harm.12

Clinical trials investigating the effect of hyperoxia on lung injury in critical care patients with or without ARDS are lacking, but high levels of FiO2 seem to have the potential to be harmful in other organs than the lung critically ill patients.18-21

Chap

ter

12269

Summary and general discussion

Prevention of lung injury in surgery patientsOne metaanalysis14 of patients without ARDS undergoing mechanical ventilation on the ICU or for surgery confirmed the findings of several clinical trials, suggesting that ventilation with lower tidal volumes decreases the occurrence of pulmonary complications and improves clinical outcomes. One problem with all these trials is that the investigators often combined the use of low tidal volumes with higher levels of PEEP,22-30 which makes it difficult if not impossible to determine the precise role of each single intervention on the beneficial effect. Some investigations suggest hyperoxia during intraoperative ventilation to have harmful effects,31, 32 but no clinical evidence on lung injury is available yet.

Summary of this thesis

The main aim of this thesis was to investigate the effect of intraoperative use of higher levels of PEEP, i.e., higher than the typical 0–2 cm H2O as applied by many anaesthesiologists, and recruitment manoeuvres on occurrence of postoperative pulmonary complications. We hypothesized that the use of higher levels of PEEP and recruitment manoeuvres would protect against development of postoperative pulmonary complications during low tidal volume ventilation. We further investigated several other aspects of perioperative ventilation, focussing on ventilation practice and the associations between ventilator settings and postoperative pulmonary complications and outcome. In this chapter we summarize the results of the studies presented in this thesis, describes how these results can be placed in context of previous research findings, and speculates on future perspectives.

In Chapter 2 we describe a metaanalysis examining the effects of intraoperative ventilator settings on postoperative outcome of non–cardiac surgery patients.33 In this analysis we tried to separate the effects of tidal volume reduction and of higher levels of PEEP on postoperative complications. We hypothesized that low tidal volumes and higher levels of PEEP with or without recruitment manoeuvres could both prevent postoperative pulmonary complications. In the analysis of eight clinical trials comprising of 1669 patients, ventilation with lower tidal volumes was associated with a lower incidence of postoperative lung injury, pulmonary infections and atelectasis compared to ventilation with conventional tidal volumes. For the examination of settings for PEEP, only five of eight trials (1323 patients) were suited for the analysis, comparing ventilation without or with lower levels of PEEP with higher levels of PEEP during surgery. Patients ventilated with higher levels of PEEP developed less postoperative lung injury and atelectasis. A beneficial effect of high levels of PEEP on postoperative pulmonary infection was also found, but in this analysis there was moderate heterogeneity. We did not find trials investigating exclusively the effects of intraoperative recruitment manoeuvres. It should be taken into account that the five trials assessing the effect of ventilation with higher levels of PEEP and recruitment manoeuvres were also part of the metaanalysis on the effect of ventilation with lower tidal volumes. In this analysis it remained uncertain whether the use of high PEEP, with or without recruitment manoeuvres, added to the beneficial effects of intraoperative use of lower tidal volumes.

270

In Chapter 3 we more extensively review the literature on intraoperative protective ventilatory strategies to prevent postoperative pulmonary complications.34 We describe six studies on predictive models of postoperative pulmonary complications, eleven trials on protective ventilation during general anaesthesia for surgery with a nonclinical primary outcome, and eight trials with a clinical primary outcome. We determined that increasing evidence shows that intraoperative lung-protective mechanical ventilation decreases the risk of postoperative pulmonary complications. However, the precise role of each single intervention remains uncertain. We conclude this review with a recommendation on intraoperative lung-protective ventilation and propose a new, alternative ventilation strategy during general anaesthesia: “permissive atelectasis”. In this new approach patients are ventilated with relatively low levels of PEEP and without recruitment manoeuvres, which reduces static stress in the lungs. Permissive atelectasis, however, could cause perioperative hypoxia, which can be counteracted by increasing in the inspired oxygen fraction.

Chapters 4 and 5 present an international prospective observational study aiming to describe current ventilator practice regarding intraoperative ventilation, and to determine any association between ventilator settings and incidence of postoperative pulmonary complications.35 We hypothesised that low tidal volumes and high levels of PEEP were commonly used, and that both are associated with a reduced incidence of postoperative pulmonary complications. During a seven–day period, data of 8,327 consecutive adult patients requiring invasive ventilation during general anaesthesia for surgery from 146 centres worldwide was collected. Intraoperative ventilation settings and postoperative data on pulmonary complications up to day 5 were collected. Our results show that patients are generally ventilated with relatively low tidal volumes (median of 500.0 [454.2 – 550.5] mL or 8.1 [7.2 – 9.1] mL/kg PBW), PEEP levels of 4.0 [0.0 – 5.0] cmH2O, and that recruitment manoeuvres are rarely performed (9.5%). The chosen level of tidal volume represents the default settings on many anaesthesia ventilators, suggesting a lack of individualisation. Postoperative pulmonary complications occurred frequently after surgery (10.4%), and were associated with longer length of hospital stay and mortality. In two different multivariate models we found that the use of higher levels of PEEP, but not size of tidal volume was independently associated with increased development of postoperative pulmonary complications.

In Chapter 6 we describe the results of an individual patient data metaanalysis using data from 15 randomized controlled trials of intraoperative ventilation.36 We tested the hypothesis that intraoperative ventilation with lower tidal volumes protects against postoperative pulmonary complications and improves clinical outcome, and that use of higher levels of PEEP adds to the beneficial effects of lower tidal volumes. The results from 2,127 patients show that intraoperative protective ventilation strategies have beneficial influence on incidence of postoperative pulmonary complications, but they did not have an effect on length of stay or mortality. Patients that developed a pulmonary complication postoperatively did have longer lengths of ICU and hospital stay and increased mortality rates. In the analysis of the individual effect of intraoperative low tidal volumes, we found decreased incidence of postoperative pulmonary complications. If higher levels of PEEP were added to low tidal volume ventilation, this did not attribute to additional benefit. The analysis on the individual effect of PEEP did not show significant influence on development of postoperative pulmonary complications.

Chap

ter

12271

Summary and general discussion

In Chapters 7, 8 and 9 we present an international trial comparing PEEP levels of 12 cmH2O combined with recruitment manoeuvres to PEEP levels of 2 cmH2O without recruitment manoeuvres in non-obese patients at high risk for postoperative pulmonary complications planned for open abdominal surgery with ventilation at tidal volumes of 8 mL/kg.37-39 In this trial we tested the hypothesis that a ventilation strategy with high levels of PEEP and recruitment manoeuvres protects against development of postoperative pulmonary complications. We randomized 900 patients from 30 centres throughout Europe and the Americas. The incidence of postoperative pulmonary complications in the first 5 days after surgery did not differ between patients receiving protective ventilation or non-protective ventilation. Patients receiving higher levels of PEEP more frequently developed intraoperative hypotension and needed more vasoactive drugs. In the lower PEEP group, the incidence of intraoperative desaturations was higher. The results of this trial indicate that protective ventilation with low tidal volumes does not gain from higher levels of PEEP with recruitment manoeuvres and may even impair hemodynamics.

In Chapter 10 we investigated whether development of postoperative lung injury attributes to incidence of morbidity and mortality, by performing a second metaanalysis using individual patient data from 12 investigations of intraoperative ventilation.40 We hypothesized that the occurrence of postoperative lung injury would be associated with worse outcome, and that postoperative outcome depends on intraoperative ventilation settings. In 3365 patients development of postoperative lung injury increased the risk of mortality, especially in patients submitted to thoracic procedures compared to abdominal procedures. The incidence of lung injury was similar between thoracic and abdominal surgery. Development of lung injury was associated with longer ICU and hospital lengths of stay. When examining the intraoperative ventilation strategies, protective ventilation was associated with lower incidence of postoperative lung injury, but not with reduced mortality rates. Also in the event of postoperative lung injury, the previously applied protective strategy of ventilation was not associated with reduced attributable mortality. This suggests that the benefits of intraoperative protective ventilation are restricted to the reduction of development of postoperative lung injury.

In Chapter 11 we extend our research from the operation room to patients ventilated in the intensive care unit after surgery. In a secondary analysis of two previous trials in patients undergoing elective and uncomplicated coronary artery bypass grafting (CABG),41, 42 we investigated the effects of PEEP on pulmonary compliance and gas exchange in the first hours of weaning from mechanical ventilation and total time on the ventilator.43 We hypothesized that higher levels of PEEP improves pulmonary function, but does not shorten duration of postoperative ventilation. We found in 121 patients that ventilation with higher levels of PEEP resulted in better compliance and oxygenation. The beneficial effect on pulmonary compliance during ventilation was only sustained when PEEP levels were kept above 5 cmH2O. After tracheal extubation the positive effect on both compliance and arterial oxygenation was not maintained. Nonetheless, patients ventilated with higher levels of PEEP required less supplemental oxygen on the ward, which could indicate that these patients had less atelectasis. Time on the ventilator was longer in patients ventilated with higher levels of PEEP. One partial explanation could be that these patients received infusion of hypnotics during a longer period of time.

272

General discussion

The main finding of this thesis is that intraoperative ventilation with higher levels of PEEP does not seem to reduce development of postoperative pulmonary complications during low tidal volume ventilation (chapter 6 and 8), in non-obese patients undergoing major surgery (cardiac, thoracic, abdominal, or spine surgery). High levels of PEEP may even be detrimental by causing hemodynamic compromise (chapter 8) and maybe even by increasing the risk of postoperative pulmonary complications (chapter 5).

In this thesis, we further show that lung-protective strategies using low tidal volumes combined with higher levels of PEEP reduce the incidence of postoperative pulmonary complications (chapters 2, 3, 6 and 10), but do not shorten hospital length of stay, or improve survival (chapters 6 and 10). In the event of a pulmonary complication, patients do have longer lengths of ICU and hospital stay, and have a higher incidence of mortality (chapter 5, 6 and 10). Finally, use of higher levels of PEEP during postoperative ventilation resulted in better compliance and oxygenation in patients undergoing elective and uncomplicated coronary artery bypass grafting, but this was not sustained after extubation and time to extubation was increased (chapter 11).

PEEP may not be beneficialContrary to general belief, this thesis shows that ventilation with higher levels of PEEP may not attribute to the protective effect of low tidal volumes on the development of postoperative pulmonary complications. Individual patient data analysis found no increased benefit of higher levels of PEEP in patients ventilated with low tidal volumes on development of pulmonary complications (chapter 6).36 Additionally, the analysis of dose–response relationship in this study suggests that there is neither a positive, nor a negative association between a higher level of PEEP and the development of postoperative pulmonary complications. In postoperative cardiac surgery patients, ventilation with higher levels of PEEP did not have a sustained effect on pulmonary compliance or arterial oxygenation (chapter 11).43 The PROVHILO trial showed that levels of 12 cmH2O PEEP did not protect against development of postoperative complications during low tidal volume ventilation (chapter 8).38 The chosen level of PEEP in this trial was criticized, because it is higher than normally applied in clinical practice.44 The resulting high end-inspiratory pressures might have caused overinflation of normally aerated alveoli. This could have diminished the clinical benefits of ventilation with higher PEEP levels. However, the level of PEEP was based on previous investigations, recommending a minimal level of 10 cmH2O PEEP to prevent cyclic opening and closing of alveoli during intraoperative ventilation.45-47 Indeed, as mentioned in our Author’s reply (chapter 9),39 the higher PEEP group showed increasing dynamic compliance during intraoperative mechanical ventilation. This suggests that effective lung recruitment was achieved, without overt overdistention. On the other hand, peak airway pressures were increased in the higher PEEP group, suggesting increased alveolar pressures that could have contributed to hyperinflation.

Experimental and clinical studies have previously questioned the beneficial effects of PEEP. As far back as the ‘70s, studies in dogs with injured lungs found that application of a level of 10 cmH2O PEEP compared to no PEEP did not counteract formation of oedema fluid, when inspiratory pressure levels were kept equal.48, 49 In pigs with non-injured lungs no difference in inflammatory

Chap

ter

12273

Summary and general discussion

response was seen between high and low levels of PEEP during low tidal ventilation.50 When examining the alveoli up-close with in vivo microscopy in pigs with surfactant deactivated lungs, alveolar stability did not differ when PEEP was set a different levels.51

In critical care patients with ARDS, no additional protective effect of higher PEEP levels was seen in mild ARDS.7 In critical care patients without ARDS, high PEEP had very limited beneficial effect on the lungs.16, 17 During intraoperative ventilation no significant beneficial effect of PEEP level above 5 cmH2O was seen on development of postoperative pulmonary complications in a retrospective study.52

PEEP may cause harmUse of higher levels of PEEP may even cause harm. High levels of PEEP can compromise intraoperative hemodynamics (chapter 8).38 Furthermore, use of PEEP may be associated with increased risk of postoperative pulmonary complications (chapter 5). In patients mechanically ventilated after CABG, high levels of PEEP prolonged weaning from the ventilator (chapter 11).43

The undesirable effect of high levels of PEEP on hemodynamics has been identified in previous investigations.53, 54 The negative effects of high levels of PEEP on development of pulmonary complications seem to be in contrast to recent clinical trials.24, 55, 56 A possible explanation could be that compared to these studies, the tidal volume size reported in chapter 5 was much smaller. Notably, the LAS VEGAS study found no association between tidal volume size and development of pulmonary complications. In the event of low tidal volume ventilation, higher levels of PEEP could attribute to hyperinflation of normally aerated lung areas and possibly augment intra-tidal shear stress of collapsed lung areas.

Earlier animal and human studies have described the potential harm of ventilation with high levels of PEEP. In healthy rats receiving low tidal volume ventilation, high levels of PEEP (15 cmH2O) caused more pulmonary oedema than levels of 0 or 10 cmH2O PEEP.57 In a surgical model of pigs without lung injury, high levels of PEEP were associated with increase of pulmonary inflammation.58 When examining different areas of rat-lung after surfactant depletion and injurious ventilation, alveolar injury was higher in the alveoli that remain open during the complete breath cycle, compared to the alveoli that collapse at end-expiration.59

In mechanically ventilated patients with acute lung injury PET-CT scans showed maximum metabolic activity in the normally aerated lung regions, and no increase in metabolic activity in regions undergoing cyclic opening and closing.60

The degree of metabolic activity in the aerated regions was associated with higher plateau pressures. This was also seen in a recent retrospective study in surgical patients, showing a strong association between reduction in plateau pressure and a decrease in postoperative pulmonary complications.52 These results suggest that even though higher levels of PEEP may lessen atelectasis formation, due to the consequential high plateau pressures normally aerated alveoli may become overdistended.

274

Driving pressureExperimental studies confirm that ventilation with higher plateau pressures can be injurious (so-called stress), but suggest that repetitive deformation during the respiratory cycle can cause more injury to the lung (so-called strain).61-64 The maximal deformation of the lung during mechanical ventilation is best expressed by calculating the driving pressure: plateau pressure minus positive end-expiratory pressure. In mechanically ventilated rats, driving pressure appeared to be one of the most important ventilation factors for lung injury.57 In studies in patients with ARDS a possible association between driving pressure and poorer outcome was identified.65-67 In a large individual patient data metaanalysis on patients with ARDS receiving protective ventilation, driving pressure was the ventilation parameter that was strongest associated with mortality.68

Functional lung sizeDriving pressure is intrinsically connected to respiratory system compliance. To inflate the lung up to tidal volume (VT), a certain pressure increase is required: the driving pressure (∆P). The amount of pressure necessary to inflate the lung depends on the compliance of the respiratory system (C = VT/∆P). Lung compliance is strongly related to the volume of the remaining aerated lung, when functional lung volume is reduced (termed functional lung size). Normalization of tidal volume to functional lung size results in driving pressure (∆P=VT/C). This is different to the generally used normalization to predicted body weight (mL/kg). The former adjusts for a possible decrease in lung aeration, while the latter corresponds with lung size of healthy, spontaneously breathing persons.

Functional lung size is decreased in patients with ARDS and in patients with healthy lungs undergoing mechanical ventilation. In ARDS, the diseased lung causes a decrease in compliance, while in patients with healthy lungs induction of anaesthesia or sedation causes large portions of the lung to collapse.69 In mechanically ventilated patients driving pressure may therefore better represent lung size, than lung size corrected for predicted body weight.

PEEP and driving pressure The effects of the level of PEEP on compliance and driving pressure are complex. In patients with decreased compliance two different phenomena may simultaneously occur when levels of PEEP are increased. First, increased levels of PEEP prevent the lung from collapsing, thereby reducing the amount of lung tissue undergoing repeated/cyclic opening and closing. This increases functional lung size and compliance, and decreases driving pressure. Second, if the higher levels of PEEP do not effectively prevent atelectasis formation, end-inspiratory pressures and transpulmonary pressures will increase and result in hyperinflation of the aerated portion of the lung.62, 69, 70 The optimum of this delicate balance in prevention of alveolar stress and strain remains unsure and quite possibly differs considerably per patient. Further investigations are warranted, to provide individually tailored mechanical ventilation guidelines for each patient.

Chap

ter

12275

Summary and general discussion

Future perspectives

Individual PEEP titrationThe findings of this thesis suggest that setting one fixed level of PEEP may not be beneficial for all surgical patients in general. An improvement could be to focus more on individualized patient care, and titrate levels of PEEP on patient-specific parameters to minimize ventilator-associated lung injury. Previous studies have titrated “best PEEP” per individual patient by searching for the highest level of PEEP that optimizes aeration and avoids cyclic opening and closing of alveoli. This thesis suggests that high levels of PEEP may be harmful to the lung. Titration of levels of PEEP should therefor focus on “lowest PEEP”, to prevent hyperinflation of patent alveoli, while avoiding severe desaturation.

Several methods have been described to titrate levels of PEEP on patient-specific physiologic parameters. Many were based on oxygenation parameters,71-76 but these correlate poorly with the amount of lung recruitment77, 78 and cannot indicate hyperinflation.

One method to titrate PEEP, is by oesophageal pressure measurement.79 By determining the oesophageal pressure, transpulmonary pressures can be calculated and used to titrate PEEP. Nonetheless, this method was primarily tested in critical care patients with ARDS. The technique is not easily transferred to the surgical population, where pneumoperitoneum and extreme positioning frequently occur and could severely alter the oesophageal pressure measurements. Furthermore, even though they seem to be strongly related, oesophageal pressures do not equal pleural pressures.80, 81

Another method is electric impedance tomography (EIT), which can image and evaluate regional distribution of alveolar aeration.82-84 With EIT the lowest levels of PEEP could be titrated by examining the aerated regions of the lung for signs of hyperinflation.85-87 Even though EIT seems to be a monitoring system with potential in experimental and clinical studies, it is not applied in clinical routine yet.

As mentioned previously, increased driving pressures seem to be strongest correlated with increased lung injury and mortality.65-68 Driving pressures may therefore be the most suitable global ventilation parameter to guide individual PEEP titration. However, the studies investigating driving pressure are all of retrospective design and performed post hoc analyses. Whether driving pressure can be manipulated at the bedside, and whether it is indeed associated with lung injury, needs to be tested in clinical trials.

Permissive atelectasisIn surgical patients with healthy lungs titration to the lowest levels of PEEP may not be necessary. Level of PEEP can be set at zero or low levels, as long as inspiratory volumes and pressures are kept within range.88, 89 In chapter 3 we proposed a new ventilation strategy termed “intraoperative permissive atelectasis”.34 This approach combines intraoperative low tidal volume ventilation with relatively low levels of PEEP without recruitment manoeuvres. If hypoxemia (SpO2 ≤ 92%) develops the FiO2 should be increased first, followed by increase of levels of PEEP, and recruitment manoeuvres based on stepwise increase of tidal volume during regular mechanical ventilation.

276

Experimental and clinical studies have shown that during ventilation with levels of low or zero PEEP inflammation is less in lung regions with atelectasis than in the normally aerated portions of the lung,51, 59-61 supporting the theory of permissive atelectasis. Permissive atelectasis could be a simple, acceptable method to reduce global strain in the lung and reduce alveolar hyperinflation during intraoperative low tidal volume ventilation in surgical patients with healthy lungs.

In conclusion

The main finding of this thesis is that intraoperative ventilation with higher levels of PEEP does not seem to reduce development of postoperative pulmonary complications during low tidal volume ventilation. Use of higher levels of PEEP may even cause harm. High levels of PEEP can compromise intraoperative hemodynamics. Furthermore, use of PEEP may be associated with increased risk of postoperative pulmonary complication.

Chap

ter

12277

Summary and general discussion

References1. Tremblay LN, Slutsky AS. Ventilator-induced lung injury: from the bench to the bedside. Intensive care medicine 2006;

32(1): 24-332. Canet J, Gallart L, Gomar C, et al. Prediction of Postoperative Pulmonary Complications in a Population-based Surgical

Cohort. Anesthesiology 2010; 113(6): 1338-503. Mazo V, Sabate S, Canet J, et al. Prospective external validation of a predictive score for postoperative pulmonary

complications. Anesthesiology 2014; 121(2): 219-314. Slutsky AS, Ranieri VM. Ventilator-induced lung injury. NEJM 2014; 370(10): 9805. Kallet RH, Matthay MA. Hyperoxic acute lung injury. Respiratory care 2013; 58(1): 123-416. Edmark L, Kostova-Aherdan K, Enlund M, Hedenstierna G. Optimal oxygen concentration during induction of general

anesthesia. Anesthesiology 2003; 98(1): 28-337. Putensen C, Theuerkauf N, Zinserling J, Wrigge H, Pelosi P. Metaanalysis: ventilation strategies and outcomes of the

acute respiratory distress syndrome and acute lung injury. Ann Intern Med 2009; 151(8): 566-768. Burns KE, Adhikari NK, Slutsky AS, et al. Pressure and volume limited ventilation for the ventilatory management of

patients with acute lung injury: a systematic review and metaanalysis. PLoS One 2011; 6(1): e146239. Meade MO, Cook DJ, Guyatt GH, et al. Ventilation strategy using low tidal volumes, recruitment maneuvers, and high

positive end-expiratory pressure for acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. JAMA 2008; 299(6): 637-45

10. Mercat A, Richard JC, Vielle B, et al. Positive end-expiratory pressure setting in adults with acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. JAMA 2008; 299(6): 646-55

11. Briel M, Meade M, Mercat A, et al. Higher vs lower positive end-expiratory pressure in patients with acute lung injury and acute respiratory distress syndrome: systematic review and metaanalysis. Jama 2010; 303(9): 865-73

12. Determann RM, Royakkers A, Wolthuis EK, et al. Ventilation with lower tidal volumes as compared with conventional tidal volumes for patients without acute lung injury: a preventive randomized controlled trial. Critical care 2010; 14(1): R1

13. Serpa Neto A, Nagtzaam L, Schultz MJ. Ventilation with lower tidal volumes for critically ill patients without the acute respiratory distress syndrome: a systematic translational review and metaanalysis. Current opinion in critical care 2014; 20(1): 25-32

14. Serpa Neto A, Cardoso SO, Manetta JA, et al. Association between use of lung-protective ventilation with lower tidal volumes and clinical outcomes among patients without acute respiratory distress syndrome: a metaanalysis. JAMA 2012; 308(16): 1651-9

15. Serpa Neto A, Simonis FD, Barbas CS, et al. Association between tidal volume size, duration of ventilation, and sedation needs in patients without acute respiratory distress syndrome: an individual patient data metaanalysis. Intensive care medicine 2014; 40(7): 950-7

16. Manzano F, Fernandez-Mondejar E, Colmenero M, et al. Positive-end expiratory pressure reduces incidence of ventilator-associated pneumonia in nonhypoxemic patients. Critical care medicine 2008; 36(8): 2225-31

17. Pepe PE, Hudson LD, Carrico CJ. Early application of positive end-expiratory pressure in patients at risk for the adult respiratory-distress syndrome. The New England journal of medicine 1984; 311(5): 281-6

18. Stub D, Smith K, Bernard S, et al. Air Versus Oxygen in ST-Segment-Elevation Myocardial Infarction. Circulation 2015; 131(24): 2143-50

19. de Jonge E, Peelen L, Keijzers PJ, et al. Association between administered oxygen, arterial partial oxygen pressure and mortality in mechanically ventilated intensive care unit patients. Critical care 2008; 12(6): R156

20. Helmerhorst HJ, Roos-Blom MJ, van Westerloo DJ, de Jonge E. Association Between Arterial Hyperoxia and Outcome in Subsets of Critical Illness: A Systematic Review, Metaanalysis, and Meta-Regression of Cohort Studies. Critical Care Medicine 2015

21. Damiani E, Adrario E, Girardis M, et al. Arterial hyperoxia and mortality in critically ill patients: a systematic review and metaanalysis. Critical care 2014; 18(6): 711

22. Maslow AD, Stafford TS, Davignon KR, Ng T. A randomized comparison of different ventilator strategies during thoracotomy for pulmonary resection. The Journal of thoracic and cardiovascular surgery 2013; 146(1): 38-44

23. Memtsoudis SG, Bombardieri AM, Ma Y, Girardi FP. The effect of low versus high tidal volume ventilation on inflammatory markers in healthy individuals undergoing posterior spine fusion in the prone position: a randomized controlled trial. Journal of clinical anesthesia 2012; 24(4): 263-9

24. Futier E, Constantin JM, Paugam-Burtz C, et al. A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. NEJM 2013; 369(5): 428-37

25. Wrigge H, Uhlig U, Zinserling J, et al. The effects of different ventilatory settings on pulmonary and systemic inflammatory responses during major surgery. Anesthesia and analgesia 2004; 98(3): 775-81.

26. Zupancich E, Paparella D, Turani F, et al. Mechanical ventilation affects inflammatory mediators in patients undergoing

278

cardiopulmonary bypass for cardiac surgery: a randomized clinical trial. The Journal of thoracic and cardiovascular surgery 2005; 130(2): 378-83

27. Reis Miranda D, Gommers D, Struijs A, et al. Ventilation according to the open lung concept attenuates pulmonary inflammatory response in cardiac surgery. Eur J Cardiothorac Surg 2005; 28(6): 889-95

28. Wolthuis EK, Choi G, Dessing MC, et al. Mechanical ventilation with lower tidal volumes and positive end-expiratory pressure prevents pulmonary inflammation in patients without preexisting lung injury. Anesthesiology 2008; 108(1): 46-54

29. Weingarten TN, Whalen FX, Warner DO, et al. Comparison of two ventilatory strategies in elderly patients undergoing major abdominal surgery. British journal of anaesthesia 2010; 104(1): 16-22

30. Lin WQ, Lu XY, Cao LH, Wen LL, Bai XH, Zhong ZJ. [Effects of the lung protective ventilatory strategy on proinflammatory cytokine release during one-lung ventilation]. Ai zheng = Aizheng = Chinese journal of cancer 2008; 27(8): 870-3

31. Meyhoff CS, Wetterslev J, Jorgensen LN, et al. Effect of high perioperative oxygen fraction on surgical site infection and pulmonary complications after abdominal surgery: the PROXI randomized clinical trial. Jama 2009; 302(14): 1543-50

32. Zoremba M, Dette F, Hunecke T, Braunecker S, Wulf H. The influence of perioperative oxygen concentration on postoperative lung function in moderately obese adults. European journal of anaesthesiology 2010; 27(6): 501-7

33. Hemmes SN, Serpa Neto A, Schultz MJ. Intraoperative ventilatory strategies to prevent postoperative pulmonary complications: a metaanalysis. Current opinion in anaesthesiology 2013; 26(2): 126-33

34. Güldner A K, MD T, Serpa Neto A, Canet J, Spieth P, Rocco PR, Schultz MJ, Pelosi P, Gama de Abreu M, for the PROtective VEntilation Network (PROVEnet) Intraoperative Protective Mechanical Ventilation for Prevention of Postoperative Pulmonary Complications - a Comprehensive Review of the Role of Tidal Volume, Positive End-Expiratory Pressure and Lung Recruitment Maneuvers. Anesthesiology 2015; 123(3): 692-713

35. Hemmes SN, de Abreu MG, Pelosi P, Schultz MJ. ESA Clinical Trials Network 2012: LAS VEGAS--Local Assessment of Ventilatory Management during General Anaesthesia for Surgery and its effects on Postoperative Pulmonary Complications: a prospective, observational, international, multicentre cohort study. European journal of anaesthesiology 2013; 30(5): 205-7

36. Serpa Neto A, Hemmes SN, Barbas CS, et al. Protective versus Conventional Ventilation for Surgery: A Systematic Review and Individual Patient Data Metaanalysis. Anesthesiology 2015, May 15 [Epub ahead of print]

37. Hemmes SN, Severgnini P, Jaber S, et al. Rationale and study design of PROVHILO - a worldwide multicenter randomized controlled trial on protective ventilation during general anesthesia for open abdominal surgery. Trials 2011; 12: 111

38. Hemmes SN, Gama de Abreu M, Pelosi P, Schultz MJ. High versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial. Lancet 2014; 384(9942): 495-503

39. Hemmes SN, de Abreu MG, Pelosi P, Schultz MJ. Positive end-expiratory pressure during surgery - Authors’ reply. Lancet 2014; 384(9955): 1670-1

40. Serpa Neto A, Hemmes SN, Barbas CS, et al. Incidence of mortality and morbidity related to postoperative lung injury in patients who have undergone abdominal or thoracic surgery: a systematic review and metaanalysis. The Lancet Respiratory medicine 2014; 2(12): 1007-15

41. Dongelmans DA, Veelo DP, Paulus F, et al. Weaning automation with adaptive support ventilation: a randomized controlled trial in cardiothoracic surgery patients. Anesthesia and analgesia 2009; 108(2): 565-71

42. Dongelmans DA, Veelo DP, Binnekade JM, et al. Adaptive support ventilation with protocolized de-escalation and escalation does not accelerate tracheal extubation of patients after nonfast-track cardiothoracic surgery. Anesthesia and analgesia 2010; 111(4): 961-7

43. Dongelmans DA, Hemmes SN, Kudoga AC, Veelo DP, Binnekade JM, Schultz MJ. Positive end-expiratory pressure following coronary artery bypass grafting. Minerva anestesiologica 2012; 78(7): 790-800

44. Futier E. Positive end-expiratory pressure in surgery: good or bad? Lancet 2014; 384(9942): 472-445. Brismar B, Hedenstierna G, Lundquist H, Strandberg A, Svensson L, Tokics L. Pulmonary densities during anesthesia

with muscular relaxation--a proposal of atelectasis. Anesthesiology 1985; 62(4): 422-846. Hachenberg T, Lundquist H, Tokics L, Brismar B, Hedenstierna G. Analysis of lung density by computed tomography

before and during general anaesthesia. Acta anaesthesiologica Scandinavica 1993; 37(6): 549-5547. Neumann P, Rothen HU, Berglund JE, Valtysson J, Magnusson A, Hedenstierna G. Positive end-expiratory pressure

prevents atelectasis during general anaesthesia even in the presence of a high inspired oxygen concentration. Acta anaesthesiologica Scandinavica 1999; 43(3): 295-301

48. Hopewell PC. Failure of positive end-expiratory pressure to decrease lung water content in alloxan-induced pulmonary edema. The American review of respiratory disease 1979; 120(4): 813-9

49. Hopewell PC, Murray JF. Effects of continuous positive-pressure ventilation in experimental pulmonary edema. J Appl Physiol 1976; 40(4): 568-74

50. Hong CM, Xu DZ, Lu Q, et al. Systemic inflammatory response does not correlate with acute lung injury associated with mechanical ventilation strategies in normal lungs. Anesthesia and analgesia 2012; 115(1): 118-21

51. Halter JM, Steinberg JM, Gatto LA, et al. Effect of positive end-expiratory pressure and tidal volume on lung injury induced by alveolar instability. Critical Care 2007; 11(1): R20

Chap

ter

12279

Summary and general discussion

52. Ladha K, Vidal Melo MF, McLean DJ, et al. Intraoperative protective mechanical ventilation and risk of postoperative respiratory complications: hospital based registry study. BMJ 2015; 351: h3646

53. Nielsen J, Ostergaard M, Kjaergaard J, et al. Lung recruitment maneuver depresses central hemodynamics in patients following cardiac surgery. Intensive care medicine 2005; 31(9): 1189-94

54. Luecke T, Pelosi P. Clinical review: Positive end-expiratory pressure and cardiac output. Critical Care 2005; 9(6): 607-2155. Ge Y, Yuan L, Jiang X, Wang X, Xu R, Ma W. [Effect of lung protection mechanical ventilation on respiratory function in

the elderly undergoing spinal fusion]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2013; 38(1): 81-556. Severgnini P, Selmo G, Lanza C, et al. Protective mechanical ventilation during general anesthesia for open abdominal

surgery improves postoperative pulmonary function. Anesthesiology 2013; 118(6): 1307-2157. Dreyfuss D, Saumon G. Role of tidal volume, FRC, and end-inspiratory volume in the development of pulmonary edema

following mechanical ventilation. The American review of respiratory disease 1993; 148(5): 1194-20358. Hong CM, Xu DZ, Lu Q, et al. Low tidal volume and high positive end-expiratory pressure mechanical ventilation

results in increased inflammation and ventilator-associated lung injury in normal lungs. Anesthesia and analgesia 2010; 110(6): 1652-60

59. Tsuchida S, Engelberts D, Peltekova V, et al. Atelectasis causes alveolar injury in nonatelectatic lung regions. American journal of respiratory and critical care medicine 2006; 174(3): 279-89

60. Bellani G, Guerra L, Musch G, et al. Lung regional metabolic activity and gas volume changes induced by tidal ventilation in patients with acute lung injury. American journal of respiratory and critical care medicine 2011; 183(9): 1193-9

61. Wakabayashi K, Wilson MR, Tatham KC, O’Dea KP, Takata M. Volutrauma, but not atelectrauma, induces systemic cytokine production by lung-marginated monocytes. Critical care medicine 2014; 42(1): e49-57

62. Caironi P, Langer T, Carlesso E, Protti A, Gattinoni L. Time to generate ventilator-induced lung injury among mammals with healthy lungs: a unifying hypothesis. Intensive care medicine 2011; 37(12): 1913-20

63. Tschumperlin DJ, Oswari J, Margulies AS. Deformation-induced injury of alveolar epithelial cells. Effect of frequency, duration, and amplitude. American journal of respiratory and critical care medicine 2000; 162(2 Pt 1): 357-62

64. Garcia CS, Rocco PR, Facchinetti LD, et al. What increases type III procollagen mRNA levels in lung tissue: stress induced by changes in force or amplitude? Respiratory physiology & neurobiology 2004; 144(1): 59-70

65. Amato MB, Barbas CS, Medeiros DM, et al. Effect of a protective-ventilation strategy on mortality in the acute respiratory distress syndrome. The New England journal of medicine 1998; 338(6): 347-54

66. Estenssoro E, Dubin A, Laffaire E, et al. Incidence, clinical course, and outcome in 217 patients with acute respiratory distress syndrome. Critical care medicine 2002; 30(11): 2450-6

67. Boissier F, Katsahian S, Razazi K, et al. Prevalence and prognosis of cor pulmonale during protective ventilation for acute respiratory distress syndrome. Intensive care medicine 2013; 39(10): 1725-33

68. Amato MB, Meade MO, Slutsky AS, et al. Driving pressure and survival in the acute respiratory distress syndrome. The New England journal of medicine 2015; 372(8): 747-55

69. Protti A, Votta E, Gattinoni L. Which is the most important strain in the pathogenesis of ventilator-induced lung injury: dynamic or static? Current opinion in critical care 2014; 20(1): 33-8

70. Gattinoni L, Carlesso E, Caironi P. Stress and strain within the lung. Current opinion in critical care 2012; 18(1): 42-771. Suter PM, Fairley B, Isenberg MD. Optimum end-expiratory airway pressure in patients with acute pulmonary failure.

The New England journal of medicine 1975; 292(6): 284-972. Stewart TE, Meade MO, Cook DJ, et al. Evaluation of a ventilation strategy to prevent barotrauma in patients at high

risk for acute respiratory distress syndrome. Pressure- and Volume-Limited Ventilation Strategy Group. The New England journal of medicine 1998; 338(6): 355-61

73. Brochard L, Roudot-Thoraval F, Roupie E, et al. Tidal volume reduction for prevention of ventilator-induced lung injury in acute respiratory distress syndrome. The Multicenter Trail Group on Tidal Volume reduction in ARDS. American journal of respiratory and critical care medicine 1998; 158(6): 1831-8

74. Brower RG, Shanholtz CB, Fessler HE, et al. Prospective, randomized, controlled clinical trial comparing traditional versus reduced tidal volume ventilation in acute respiratory distress syndrome patients. Critical care medicine 1999; 27(8): 1492-8

75. Murray IP, Modell JH, Gallagher TJ, Banner MJ. Titration of PEEP by the arterial minus end-tidal carbon dioxide gradient. Chest 1984; 85(1): 100-4

76. Downs JB, Klein EF, Jr., Modell JH. The effect of incremental PEEP on PaO 2 in patients with respiratory failure. Anesthesia and analgesia 1973; 52(2): 210-5

77. Dantzker DR, Lynch JP, Weg JG. Depression of cardiac output is a mechanism of shunt reduction in the therapy of acute respiratory failure. Chest 1980; 77(5): 636-42

78. Cressoni M, Caironi P, Polli F, et al. Anatomical and functional intrapulmonary shunt in acute respiratory distress syndrome. Critical care medicine 2008; 36(3): 669-75

79. Talmor D, Sarge T, Malhotra A, et al. Mechanical ventilation guided by esophageal pressure in acute lung injury. The New England journal of medicine 2008; 359(20): 2095-104

280

80. Talmor D, Sarge T, O’Donnell CR, et al. Esophageal and transpulmonary pressures in acute respiratory failure. Critical care medicine 2006; 34(5): 1389-94

81. Hager DN, Brower RG. Customizing lung-protective mechanical ventilation strategies. Critical care medicine 2006; 34(5): 1554-5

82. Costa EL, Borges JB, Melo A, et al. Bedside estimation of recruitable alveolar collapse and hyperdistension by electrical impedance tomography. Intensive care medicine 2009; 35(6): 1132-7

83. Meier T, Luepschen H, Karsten J, et al. Assessment of regional lung recruitment and derecruitment during a PEEP trial based on electrical impedance tomography. Intensive care medicine 2008; 34(3): 543-50

84. Luepschen H, Meier T, Grossherr M, Leibecke T, Karsten J, Leonhardt S. Protective ventilation using electrical impedance tomography. Physiological measurement 2007; 28(7): S247-60

85. Zhao Z, Steinmann D, Frerichs I, Guttmann J, Moller K. PEEP titration guided by ventilation homogeneity: a feasibility study using electrical impedance tomography. Critical care 2010; 14(1): R8

86. Karsten J, Grusnick C, Paarmann H, Heringlake M, Heinze H. Positive end-expiratory pressure titration at bedside using electrical impedance tomography in post-operative cardiac surgery patients. Acta anaesthesiologica Scandinavica 2015; 59(6): 723-32

87. Blankman P, Hasan D, Erik G, Gommers D. Detection of ‘best’ positive end-expiratory pressure derived from electrical impedance tomography parameters during a decremental positive end-expiratory pressure trial. Critical Care 2014; 18(3): R95

88. Eikermann M, Kurth T. Apply Protective Mechanical Ventilation in the Operating Room in an Individualized Approach to Perioperative Respiratory Care. Anesthesiology 2015

89. Pelosi P, Ravagnan I, Giurati G, et al. Positive end-expiratory pressure improves respiratory function in obese but not in normal subjects during anesthesia and paralysis. Anesthesiology 1999; 91(5): 1221-31

Chap

ter

12281

Summary and general discussion

Appendices

284

Nederlandse Samenvatting

Introductie

Postoperatieve pulmonale complicatiesTijdens en na operaties wordt de ademhaling van patiënten vaak tijdelijk geregeld middels een beademingsmachine (mechanische ventilatie). De belangrijkste instellingen van de beademingsmachine hebben betrekking op de druk waarmee lucht wordt ingeblazen en het volume dat wordt ingeblazen. Suboptimale instellingen kunnen schade aan de long veroorzaken.1 Dit vergroot waarschijnlijk het risico op het ontwikkelen van postoperatieve complicaties met betrekking tot de longen2 en kan het klinisch herstel doen verslechteren.3 Toch is het niet geheel duidelijk wat precies de beste instellingen zijn.

Longschade door mechanische ventilatieEr zijn meerdere mechanismen beschreven die tijdens mechanische beademing de long kunnen beschadigen. De twee belangrijkste mechanismen zijn het overrekken van longblaasjes aan het einde van de inademing en het herhaaldelijk openen en sluiten van longblaasjes die samenvallen tijdens de uitademing.4 Het gebruik van te hoge drukken tijdens de inademing of te grote volumes bij een ademteug (teugvolumes), kan overrekking van longblaasjes veroorzaken. Dit kan verergerd worden door te hoge drukken aan het einde van de uitademing (positive end–expiratory pressure, PEEP). Echter, wanneer de PEEP drukken te laag zijn, kan dit resulteren in herhaaldelijk openen en sluiten van longblaasjes tijdens de ademcyclus. Naast deze ‘mechanische stress’ ondergaat de long ook ‘chemische stress’ tijdens mechanische beademing. Een hoog percentage zuurstof (FiO2) in de inademingslucht verhoogt de aanmaak van zogenaamde reactieve zuurstofradicalen, wat schadelijk is voor de cellen.5 Een hoog percentage FiO2 (> 0.8) verergert tevens het samenvallen van longblaasjes (atelectase), omdat zuurstof heel snel wordt opgenomen door het lichaam.6 Op grond van deze mechanismen, bestaat long-beschermende beademing uit beademing met lage teugvolumes om overrekking van longblaasjes te voorkomen en met positieve drukken aan het einde van de expiratie (PEEP) om repetitief openen en sluiten van longblaasjes te vermijden. Hier kunnen nog zogenaamde rekruteer manouvres aan worden toegevoegd. Met deze techniek wordt geprobeerd samengevallen longblaasjes weer te openen. Lagere concentraties zuurstof zouden daarbij kunnen beschermen tegen longschade door het voorkomen van een te hoog zuurstofgehalte in de longen en het bloed.

Long-beschermende beademing van patiënten op de Intensive CareBij patiënten met acute respiratory distress syndrome of ARDS (een ernstige longaandoening dat gekenmerkt wordt door een heftige ontstekingsreactie in de longen) is uit gerandomiseerde onderzoeken gebleken dat lage teugbeademing de incidentie van longschade en zelfs sterfte vermindert. Dit is bevestigd door twee grote metaanalyses.7, 8 Beademing met hogere PEEP niveaus liet in drie verschillende trials geen beschermend effect zien bij patiënten met ARDS,9-

11 terwijl op grond van een grote meta-analyse hogere PEEP niveaus wel bescherming lijken te bieden bij patiënten met matige of ernstige ARDS.7

Bij patiënten op de Intensive Care zonder ARDS is het bewijs voor het beschermende effect van lage teugen minder sterk. De auteurs van een gerandomiseerde trial concluderen dat het

285

Nederlandse samenvatting

gebruik van lage teugvolumes resulteert in een lagere incidentie van longschade.12 Dit is bevestigd door een conventionele meta-analyse13 en twee metaanalyses waarbij individuele patiëntdata werden gebruikt.14, 15 De geïncludeerde studies in deze metaanalyses waren echter van matige kwaliteit, waardoor de uitkomsten met enige voorzichtigheid dienen te worden geïnterpreteerd. Voor het gebruik van PEEP bij patiënten op de Intensive Care zonder ARDS is weinig onderzoek beschikbaar en worden tegenstrijdige resultaten gerapporteerd. De resultaten van een trial suggereerden een beschermend effect van hogere PEEP niveaus,16, 17 in een trial werd geen effect gevonden12 en de auteurs van een trial stelden zelfs nadelige effecten vast.12 Er zijn geen klinische trials beschikbaar met de schadelijke effecten van een hoog percentage O2 op de longen als onderwerp. Een hoog percentage O2 lijkt wel schade te kunnen veroorzaken in andere organen, bij beademende patiënten met of zonder ARDS op de Intensive Care.18-21

Long-beschermende beademing in chirurgische patiënten Het potentieel beschermende effect van long-protectieve beademing op de Intensive Care wakkerde interesse aan voor long-beschermende beademing ook toe te passen rondom operaties. De laatste jaren zijn verschillende studies naar beademing van chirurgische patiënten op de operatiekamer verricht. Studies naar lage teugbeademing, stelden vast dat de incidentie van postoperatieve complicaties daalde en het klinisch beloop verbeterde.22-24 Andere studies combineerden lage teugen met hogere PEEP niveaus en vonden ook een verbeterde uitkomst.25-33 Een grote meta-analyse van studies naar long-beschermende beademing in patiënten zonder longschade die beademend werden op de Intensive Care of tijdens chirurgie bevestigt deze resultaten.14 Bij veel van deze studies is het echter moeilijk om te onderscheiden welk element het beschermende effect veroorzaakte: lage teugen of hogere PEEP niveaus, of rekruteer manoeuvres, of een combinatie van deze drie. Klinische studies naar het schadelijke effect van een hoog zuurstofpercentage op de longen zijn niet beschikbaar, maar ook in deze patiënten lijken hoge zuurstoffracties schadelijk.34, 35

Samenvatting van dit proefschrift

Doelen proefschriftDit proefschrift, genaamd long-beschermende beademing rondom en tijdens operaties, richt zich op verschillende aspecten van beademing tijdens en na een operatie, met name op de klinische praktijk en de associaties tussen verschillende beademingsinstellingen en postoperatieve complicaties van de longen. De focus hierbij ligt vooral op PEEP, waarbij wij het effect van intra-operatieve beademing met hogere PEEP niveaus en rekruteer manoeuvres op ontwikkelen van postoperatieve pulmonale complicaties tijdens lage teugbeademing gedurende open buik chirurgie onderzochten. Onze hypothese was dat het gebruik van hogere PEEP niveaus met rekruteer manoeuvres zou beschermen tegen het ontwikkelen van postoperatieve pulmonale complicaties.

SamenvattingHoofdstuk 1 bestaat uit een algemene introductie, waarin de literatuur gepubliceerd voor het uitvoeren van de studies in dit proefschrift wordt samengevat. Hoofdstuk 2 bevat een meta-

286

analyse van acht klinische studies die de effecten van intra-operatieve beademingsinstellingen op postoperatieve uitkomsten vaststelden bij patiënten die niet-cardiale chirurgie ondergingen.36 Hierbij poogden wij onderscheid te maken tussen de effecten van lage teugen en van hogere PEEP niveaus. Onze hypothese was dat zowel lage teugen als hogere PEEP niveaus met of zonder rekruteer manoeuvres het ontwikkelen van postoperatieve pulmonale complicaties kon voorkomen. In een populatie van 1669 patiënten was beademing met lage teugen geassocieerd met minder postoperatieve long schade, longontsteking en atelectase, vergeleken met conventionele beademing. In de meta-analyse met als onderwerp PEEP, konden maar vijf van de acht onderzoeken worden meegenomen (1323 patiënten). In deze vijf onderzoeken werden lage PEEP niveaus (0 tot 3 cmH2O) met hogere PEEP niveaus (3 – 12 cmH2O) vergeleken. Patiënten die met hogere PEEP niveaus werden beademend ontwikkelden minder postoperatieve longschade en atelectase. Daarbij hadden zij minder vaak een longontsteking, al was er sprake van enige heterogeniteit in deze analyse. Er waren geen trials beschikbaar die specifiek rekruteer manoeuvres onderzochten. Een punt van aandacht is dat de vijf studies naar PEEP niveaus in de meta-analyse ook deel waren van de meta-analyse naar lage teugvolumes. Daardoor blijft het in deze analyse onzeker of hogere PEEP niveaus met of zonder rekruteer manoeuvres bijdragen aan het beschermde effect van lage teugbeademing.

Hoofdstuk 3 bevat een uitgebreider overzicht van de literatuur over intra-operatieve beschermende beademing.37 Wij beschrijven hierin zes studies naar preoperatieve voorspellende modellen van postoperatieve pulmonale complicaties, 11 studies naar beschermende beademing tijdens algehele anesthesie met een niet-klinisch primair eindpunt en acht studies naar beschermende beademing met een klinisch primair eindpunt. Uit deze studies concluderen wij dat er toenemend bewijs is dat intra-operatieve long-beschermende beademing het risico op postoperatieve pulmonale complicaties vermindert. De exacte bijdrage van de afzonderlijke elementen (teugvolume, PEEP en rekruteer manouvres) bij long-beschermende beademing blijft echter onduidelijk. Wij sluiten het overzicht af met een voorstel voor een nieuwe benadering van intra-operatieve long-beschermende beademing: ‘permissive atelectasis’, oftewel ‘geaccepteerd samenvallen van longdelen’. In deze nieuwe strategie worden patiënten beademend met relatief lage PEEP niveaus zonder rekruteer manoeuvres, wat waarschijnlijk overrekking van longblaasjes vermindert. ‘Permissive atelectasis’ kan echter wel leiden tot lage zuurstoffracties en een lage zuurstofsaturatie in het bloed. Dit kan eventueel worden opgevangen door het percentage ingeademde zuurstof licht te verhogen.

In hoofdstuk 4 en 5 presenteren wij een internationale prospectieve observationele studie, gericht op het in kaart brengen van de huidige intra-operatieve beademingsstrategieën en op het onderzoeken de relatie tussen verschillende beademingsinstellingen en het optreden van postoperatieve pulmonale complicaties.38, Onze hypothese was dat lage teugvolumes en hogere PEEP niveaus het meest toegepast worden, en dat beiden een associatie hebben met een verminderde incidentie van postoperatieve pulmonale complicaties. Gedurende een periode van zeven dagen werd in 146 centra wereldwijd data verzameld van 8,327 volwassen chirurgische patiënten die mechanische beademing tijdens algehele anesthesie ondergingen. Intra-operatieve beademingsinstellingen werden verzameld en postoperatieve pulmonale complicaties gedurende de eerste 5 postoperatieve dagen werden geregistreerd. Onze resultaten laten zien dat patiënten over het algemeen worden beademd met relatief lage teugvolumes (mediaan van

287

Nederlandse samenvatting

500.0 [454.2 – 550.5] mL of 8.1 [7.2 – 9.1] mL/kg PBW), een PEEP niveau van 4.0 [0.0 – 5.0] cmH2O en dat rekruteer manoeuvres zelden worden toegepast (9.5%). De maat van de gekozen teugvolumes lijkt sterk op de standard instellingen van veel beademingsmachines (500 mL), wat doet vermoeden dat de beademingsinstellingen weinig op het individu worden aangepast. Postoperatieve pulmonale complicaties kwamen vaak voor (10.4%) en waren geassocieerd met langere ziekenhuisopname en hogere ziekenhuismortaliteit. Twee methodologisch verschillende multivariate analyses lieten beiden zien dat hogere PEEP niveaus geassocieerd waren met het ontwikkelen van postoperatieve pulmonale complicaties, terwijl dit niet gold voor grotere teugvolumes.

In hoofdstuk 6 beschrijven wij de resultaten van een meta-analyse van individuele patiëntdata van 15 gerandomiseerde studies naar intra-operatieve long-beschermende beademing.39 Onze hypothese was dat intra-operatieve beademing met lage teugvolumes beschermt tegen het ontwikkelen van postoperatieve pulmonale complicaties en het klinisch beloop (duur van opname en ziekenhuissterfte) verbetert. Verder was onze hypothese dat het gebruik van hogere PEEP niveaus bijdraagt aan het beschermende effect van lage teugvolumes. De resultaten (verkregen bij totaal 2,127 patiënten) lieten zien dat intra-operatieve long-beschermende beademing de incidentie van postoperatieve pulmonale complicaties vermindert, maar dat het geen effect heeft op het verdere postoperatieve beloop. Patiënten die een pulmonale complicatie ontwikkelden in de postoperatieve periode hadden wel een verlengde duur van opname op de Intensive Care en in het ziekenhuis, alsmede een verhoogde mortaliteit. De analyse van het individuele effect van lage teugvolumes, liet een verminderde incidentie van postoperatieve pulmonale complicaties zien. Wanneer het gebruik van hogere PEEP niveaus werd toegevoegd aan de analyse van lage teugvolumes, nam het beschermende effect niet toe. In de analyse van het individuele effect van hogere PEEP niveaus werd geen significant effect op het ontwikkelen van postoperatieve pulmonale complicaties gezien.

In hoofdstukken 7 en 8 beschrijven wij een internationale studies waarin beademing met PEEP niveaus van 12 cmH2O gecombineerd met rekruteer manoeuvres worden vergeleken met PEEP niveaus van 2 cmH2O zonder rekruteer manoeuvres tijdens beademing met lage teugvolumes in niet-obese patiënten met een verhoogd risico op het ontwikkelen van postoperatieve pulmonale complicaties die open buik chirurgie ondergaan.40, 41 In deze studie hebben wij de hypothese getest of beademing met hogere PEEP niveaus met rekruteer manoeuvres beschermt tegen het ontwikkelen van postoperatieve pulmonale complicaties. Wij hebben 900 patiënten uit 30 centra verspreid over Europa en Noord- en Zuid-Amerika geïncludeerd. De incidentie van pulmonale complicaties gedurende de eerste 5 postoperatieve dagen verschilde niet tussen patiënten die hogere PEEP niveaus ontvingen en patiënten die met lage PEEP niveaus werden beademend. Patiënten beademd met hogere PEEP niveaus ontwikkelden vaker lage bloeddrukken tijdens de operatie en hadden vaker medicatie nodig om de bloeddruk te verhogen. Patiënten beademd met lage PEEP niveaus hadden intra-operatief vaker een lage zuurstofsaturatie. De resultaten van deze trial suggereren dat long-beschermende beademing met lage teugvolumes niet verder verbeterd wordt door het toepassen van hogere PEEP niveaus met rekruteer manoeuvres tijdens intra-operatieve beademing en dat hogere PEEP niveaus mogelijk kunnen leiden tot een verslechterde hemodynamiek. In hoofdstuk 9 presenteren wij enkele commentaren op dit stuk, samen met onze reactie hierop.42

288

In de studie gerapporteerd in hoofdstuk 10 hebben wij onderzocht of het ontwikkelen van postoperatieve longschade bijdraagt aan sterfte. Wij hebben een tweede meta-analyse gedaan waarbij we individuele patiëntdata uit 12 verschillende onderzoeken naar intra-operatieve beschermende beademing hebben gebruikt.43 Onze hypothese was dat het ontwikkelen van postoperatieve longschade geassocieerd is met een slechtere uitkomst en dat de intra-operatieve beademingsinstellingen effect hebben op het postoperatieve beloop. In een cohort van 3365 patiënten bleek het ontwikkelen van postoperatieve longschade geassocieerd te zijn met een verhoogde mortaliteit, met name bij patiënten die geopereerd werden in de borstkas (thoracale procedure), vergeleken met patiënten die buikchirurgie ondergingen. Het ontwikkelen van postoperatieve longschade kwam tussen deze twee groepen overeen. Het ontstaan van longschade was geassocieerd met een langere opnameduur op de Intensive Care en in het ziekenhuis. Het gebruik van beschermende beademingsstrategieën was geassocieerd met een lagere incidentie van postoperatieve longschade, maar het had geen effect op het klinisch beloop. Wanneer een patiënt postoperatief longschade ontwikkelde bleek de voorafgaande intra-operatieve beademing geen invloed te hebben op het wel of niet overlijden van de patiënt. Dit suggereert dat de positieve invloed van intra-operatieve long-beschermende beademing beperkt is tot het verminderen van postoperatieve longschade en geen invloed heeft op mortaliteit.

In hoofdstuk 11 beschrijven wij hoe we ons onderzoek uitbreidden van de operatiekamers naar patiënten die postoperatief beademd worden op de Intensive Care. Hiervoor analyseerden wij data van twee eerdere studies naar patiënten die een geplande ongecompliceerde coronaire bypass operatie ondergingen.44, 45 Wij onderzochten of beademen met hogere PEEP niveaus tijdens de postoperatieve periode effect had op de compliantie van het respiratoire systeem en op gasuitwisseling in de long tijdens de eerste uren postoperatieve beademing. Met de compliantie van het respiratoire systeem wordt de verandering van longvolume bij een bepaalde beademingsdruk bedoeld, dit komt overeen met de elastische krachten van de long. Verder onderzochten wij of beademen met hogere PEEP niveaus invloed had op de totale duur van postoperatieve beademing.46 Wij testten de hypothese dat hogere PEEP niveaus resulteren in betere compliantie en gaswisseling, maar geen invloed hebben op duur van postoperatieve beademing. In een cohort van 121 patiënten stelden wij vast dat hogere PEEP niveaus leidden tot betere compliantie en gaswisseling, maar dat dit effect alleen bleef bestaan bij PEEP niveaus boven 5 cmH2O. Na verwijderen van de beademingsbuis verdwenen de verbeterde compliantie en gaswisseling. Patiënten die beademend werden met hogere PEEP niveaus hadden minder extra zuurstoftoediening nodig op de afdeling waar ze verder herstelden van hun operatie, wat suggereert dat zij mogelijk minder vorming van atelectase hadden. De totale duur van de postoperatieve beademing was langer in patiënten beademd met hogere PEEP niveaus. Een gedeeltelijke verklaring hiervoor zou kunnen zijn dat deze patiënten meer en langer slaapmiddelen kregen toegediend.

Discussie

De belangrijkste bevinding van dit proefschrift is dat intra-operatieve beademing met hogere PEEP niveaus in patiënten met een normaal gewicht die een grote chirurgische ingreep ondergaan niet lijkt te beschermen tegen het ontwikkelen van postoperatieve pulmonale complicaties

289

Nederlandse samenvatting

tijdens beademing met lage teugvolumes (hoofdstuk 6 en 8). Hogere PEEP niveaus kunnen zelfs potentieel schade veroorzaken, door verslechtering van de hemodynamiek (hoofdstuk 8) en mogelijk ook door toename van het risico op postoperatieve pulmonale complicaties (hoofdstuk 5).

In dit proefschrift laten wij verder zien dat long-protectieve beademing met lage teugen en hogere PEEP niveaus beschermt tegen het ontwikkelen van postoperatieve pulmonale complicaties, maar dat dit geen invloed heeft op duur van opname of mortaliteit (hoofdstukken 6 en 10). Patiënten die postoperatief een pulmonale complicatie ontwikkelen hebben wel een langere opnameduur en een hogere mortaliteit (hoofdstukken 5, 6 en 10). Tot slot, bij patiënten die een cardiale bypass operatie ondergingen verbetert het toedienen van hogere PEEP niveaus tijdens postoperatieve beademing de compliantie en oxygenatie, maar dit houdt geen stand na het stoppen van de beademing en het verwijderen van de beademingsbuis. De duur tussen het einde van de operatie en extubatie was in deze groep verlengd (hoofdstuk 11).

PEEP beschermt mogelijk nietIn tegenstelling tot ‘de algemene opvatting’, laat dit proefschrift zien dat beademing met hogere PEEP niveaus niet bijdraagt aan het beschermende effect van beademing met lage teugen op het ontwikkelen van postoperatieve pulmonale complicaties. Een meta-analyse van individuele patiëntdata liet zien dat er geen toegevoegd beschermend effect van hogere PEEP niveaus is op de incidentie van pulmonale complicaties in patiënten die beademend werden met lage teugvolumes (hoofdstuk 6).39 Dezelfde studie vond geen directe associatie tussen het niveau van PEEP en het ontwikkelen van postoperatieve pulmonale complicaties. Bij patiënten na een cardiale bypass operatie bleek beademing met hogere PEEP niveaus geen blijvend effect te hebben op longcompliantie of verbeterde gaswisseling (hoofdstuk 11).46 Uit de PROVHILO trial bleek dat beademing met PEEP van 12 cmH2O niet beschermde tegen het ontwikkelen van postoperatieve complicaties tijdens beademing met lage teugen (hoofdstuk 8).41 Het niveau van PEEP dat gebruikt werd in deze studie werd bekritiseerd, omdat het hoger is dan het niveau van PEEP dat over het algemeen wordt toegepast in de klinische praktijk.47 De hogere PEEP drukken zouden de drukken aan het eind van de inspiratie hebben verhoogd, wat mogelijk de longblaasjes heeft kunnen overrekken. Dit zou het beschermende effect van hogere niveaus van PEEP teniet kunnen hebben gedaan. Wij hebben echter het niveau van PEEP gebaseerd op eerdere experimenten, waar een PEEP niveau boven de 10 cmH2O werd aangeraden om het repetitief openen en sluiten van de longblaasjes tijdens intra-operatieve beademing te voorkomen.48-50 Zoals wij bespreken in onze ‘Author’s reply’ (hoofdstuk 9),42 hebben wij in onze studie geobserveerd dat de hogere PEEP groep een toename van compliantie had tijdens intra-operatieve beademing. Dit suggereert dat de longblaasjes effectief worden geopend tijdens de beademing, zonder duidelijke tekenen van overrekking. Aan de andere kant zagen wij ook dat de hogere PEEP groep hogere drukken aan het einde van de inademing had (zogenaamde peak drukken), wat bijgedragen zou kunnen hebben aan overrekking van delen van de long.

PEEP kan mogelijk schade veroorzakenHet gebruik van hogere PEEP niveaus zou zelfs mogelijk schade kunnen veroorzaken. Hogere PEEP niveaus verslechteren de intra-operatieve hemodynamiek (hoofdstuk 8)41 en kunnen zelfs het risico op postoperatieve pulmonale complicaties verhogen (hoofdstuk 5). Bij beademde

290

patiënten na cardiale bypass chirurgie lijken hogere PEEP niveaus de totale postoperatieve beademingsduur te verlengen (hoofdstuk 11).46

Het nadelige effect van PEEP op de hemodynamiek is eerder beschreven.51, 52 De negatieve effecten van PEEP op het ontwikkelen van postoperatieve pulmonale complicaties is echter in strijd met eerdere recente studies.27, 53, 54 Een mogelijke verklaring hiervoor is, dat vergeleken met deze trials, de toegediende teugvolumes veel kleiner waren. In de LAS VEGAS studie vonden wij ook geen verband tussen de grootte van teugvolumes en het ontwikkelen van postoperatieve complicaties. Wanneer beademd wordt met lage teugvolumes, zouden te hoge PEEP niveaus mogelijk kunnen bijdragen aan het overrekken van bepaalde longdelen.

Driving pressureExperimentele studies laten inderdaad zien dat beademing met te hoge drukken aan het einde van de inademing (plateau drukken) schadelijk is voor de long (‘stress’). Echter, mogelijk is de repetitieve uitrekking van longblaasjes gedurende de ademcyclus (‘strain’) meer schadelijk.55-58 De maximale uitrekking van de long tijdens beademing kan het beste worden berekend door de ‘driving pressure’: de drukken bij het einde van de inademing (plateau drukken) minus de positieve drukken bij het einde van de uitademing (PEEP).

In beademde ratten lijkt driving pressure de belangrijkste schadelijke factor van beademing.59 In studies bij patiënten met ARDS lijkt er een associatie te bestaan tussen driving pressure en slechtere klinische uitkomst.60-62 In een grote individuele patiëntdata analyse van klinische studies die het effect van beschermende beademing onderzochten in patiënten met ARDS, bleek driving pressure van alle beademingsfactoren het sterkst geassocieerd met mortaliteit.63

PEEP en driving pressure De effecten van PEEP op het open houden van de long versus het overrekken van de long is complex. Aan de ene kant kan PEEP voorkomen dat longdelen samenvallen, waardoor de long homogeen open blijft en het repetitieve samenvallen van longdelen aan het einde van de uitademing wordt voorkomen. Aan de andere kant, wanneer het niveau van PEEP niet effectief de long open houdt, stijgen de drukken aan het einde van de inademing, wat kan leiden tot overrekking van longblaasjes en tot schade aan de long.56, 64, 65 Het optimum binnen dit delicate balans blijft onduidelijk en verschilt mogelijk per patiënt.

Wellicht kan PEEP in de toekomst per patiënt ingesteld worden, zodat de optimale driving pressure (plateau drukken min PEEP) kan worden gevonden per patiënt en zo longschade kan worden verminderd.

Meer onderzoek zal moeten worden verricht naar de effecten van driving pressure op postoperatieve complicaties en hoe driving pressure (en dus PEEP) het beste kan worden ingesteld op de beademingsmachine.

291

Nederlandse samenvatting

Conclusie

Dit proefschrift toont als eerste aan dat intra-operatieve beademing met hogere PEEP niveaus in patiënten met een normaal gewicht die een grote chirurgische ingreep ondergaan niet lijkt te beschermen tegen het ontwikkelen van postoperatieve pulmonale complicaties tijdens beademing met lage teugvolumes. Hogere PEEP niveaus kunnen zelfs mogelijk schade veroorzaken door verslechtering van de intra-operatieve hemodynamiek en toename van het risico op postoperatieve pulmonale complicaties.

292

Referenties1. Tremblay LN, Slutsky AS. Ventilator-induced lung injury: from the bench to the bedside. Intensive care medicine 2006;

32(1): 24-332. Canet J, Gallart L, Gomar C, et al. Prediction of Postoperative Pulmonary Complications in a Population-based Surgical

Cohort. Anesthesiology 2010; 113(6): 1338-503. Mazo V, Sabate S, Canet J, et al. Prospective external validation of a predictive score for postoperative pulmonary

complications. Anesthesiology 2014; 121(2): 219-314. Slutsky AS, Ranieri VM. Ventilator-induced lung injury. The New England journal of medicine 2014; 370(10): 9805. Kallet RH, Matthay MA. Hyperoxic acute lung injury. Respiratory care 2013; 58(1): 123-416. Edmark L, Kostova-Aherdan K, Enlund M, Hedenstierna G. Optimal oxygen concentration during induction of general

anesthesia. Anesthesiology 2003; 98(1): 28-337. Putensen C, Theuerkauf N, Zinserling J, Wrigge H, Pelosi P. Metaanalysis: ventilation strategies and outcomes of the

acute respiratory distress syndrome and acute lung injury. Ann Intern Med 2009; 151(8): 566-768. Burns KE, Adhikari NK, Slutsky AS, et al. Pressure and volume limited ventilation for the ventilatory management of

patients with acute lung injury: a systematic review and metaanalysis. PLoS One 2011; 6(1): e146239. Meade MO, Cook DJ, Guyatt GH, et al. Ventilation strategy using low tidal volumes, recruitment maneuvers, and high

positive end-expiratory pressure for acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. Jama 2008; 299(6): 637-45

10. Mercat A, Richard JC, Vielle B, et al. Positive end-expiratory pressure setting in adults with acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. Jama 2008; 299(6): 646-55

11. Briel M, Meade M, Mercat A, et al. Higher vs lower positive end-expiratory pressure in patients with acute lung injury and acute respiratory distress syndrome: systematic review and metaanalysis. Jama 2010; 303(9): 865-73

12. Determann RM, Royakkers A, Wolthuis EK, et al. Ventilation with lower tidal volumes as compared with conventional tidal volumes for patients without acute lung injury: a preventive randomized controlled trial. Critical care 2010; 14(1): R1

13. Serpa Neto A, Nagtzaam L, Schultz MJ. Ventilation with lower tidal volumes for critically ill patients without the acute respiratory distress syndrome: a systematic translational review and metaanalysis. Current opinion in critical care 2014; 20(1): 25-32

14. Serpa Neto A, Cardoso SO, Manetta JA, et al. Association between use of lung-protective ventilation with lower tidal volumes and clinical outcomes among patients without acute respiratory distress syndrome: a metaanalysis. Jama 2012; 308(16): 1651-9

15. Serpa Neto A, Simonis FD, Barbas CS, et al. Association between tidal volume size, duration of ventilation, and sedation needs in patients without acute respiratory distress syndrome: an individual patient data metaanalysis. Intensive care medicine 2014; 40(7): 950-7

16. Pepe PE, Hudson LD, Carrico CJ. Early application of positive end-expiratory pressure in patients at risk for the adult respiratory-distress syndrome. The New England journal of medicine 1984; 311(5): 281-6

17. Manzano F, Fernandez-Mondejar E, Colmenero M, et al. Positive-end expiratory pressure reduces incidence of ventilator-associated pneumonia in nonhypoxemic patients. Critical care medicine 2008; 36(8): 2225-31

18. Stub D, Smith K, Bernard S, et al. Air Versus Oxygen in ST-Segment-Elevation Myocardial Infarction. Circulation 2015; 131(24): 2143-50

19. de Jonge E, Peelen L, Keijzers PJ, et al. Association between administered oxygen, arterial partial oxygen pressure and mortality in mechanically ventilated intensive care unit patients. Critical care 2008; 12(6): R156

20. Helmerhorst HJ, Roos-Blom MJ, van Westerloo DJ, de Jonge E. Association Between Arterial Hyperoxia and Outcome in Subsets of Critical Illness: A Systematic Review, Metaanalysis, and Meta-Regression of Cohort Studies. Critical care medicine 2015

21. Damiani E, Adrario E, Girardis M, et al. Arterial hyperoxia and mortality in critically ill patients: a systematic review and metaanalysis. Critical care 2014; 18(6): 711

22. Schilling T, Kozian A, Huth C, et al. The pulmonary immune effects of mechanical ventilation in patients undergoing thoracic surgery. Anesthesia and analgesia 2005; 101(4): 957-65

23. Sundar S, Novack V, Jervis K, et al. Influence of low tidal volume ventilation on time to extubation in cardiac surgical patients. Anesthesiology 2011; 114(5): 1102-10

24. Treschan TA, Kaisers W, Schaefer MS, et al. Ventilation with low tidal volumes during upper abdominal surgery does not improve postoperative lung function. British journal of anaesthesia 2012; 109(2): 263-71\

25. Maslow AD, Stafford TS, Davignon KR, Ng T. A randomized comparison of different ventilator strategies during thoracotomy for pulmonary resection. The Journal of thoracic and cardiovascular surgery 2013; 146(1): 38-44

26. Memtsoudis SG, Bombardieri AM, Ma Y, Girardi FP. The effect of low versus high tidal volume ventilation on inflammatory

293

Nederlandse samenvatting

markers in healthy individuals undergoing posterior spine fusion in the prone position: a randomized controlled trial. Journal of clinical anesthesia 2012; 24(4): 263-9

27. Futier E, Constantin JM, Paugam-Burtz C, et al. A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. The New England journal of medicine 2013; 369(5): 428-37

28. Wrigge H, Uhlig U, Zinserling J, et al. The effects of different ventilatory settings on pulmonary and systemic inflammatory responses during major surgery. Anesthesia and analgesia 2004; 98(3): 775-81

29. Zupancich E, Paparella D, Turani F, et al. Mechanical ventilation affects inflammatory mediators in patients undergoing cardiopulmonary bypass for cardiac surgery: a randomized clinical trial. The Journal of thoracic and cardiovascular surgery 2005; 130(2): 378-83

30. Reis Miranda D, Gommers D, Struijs A, et al. Ventilation according to the open lung concept attenuates pulmonary inflammatory response in cardiac surgery. Eur J Cardiothorac Surg 2005; 28(6): 889-95

31. Wolthuis EK, Choi G, Dessing MC, et al. Mechanical ventilation with lower tidal volumes and positive end-expiratory pressure prevents pulmonary inflammation in patients without preexisting lung injury. Anesthesiology 2008; 108(1): 46-54

32. Weingarten TN, Whalen FX, Warner DO, et al. Comparison of two ventilatory strategies in elderly patients undergoing major abdominal surgery. British journal of anaesthesia 2010; 104(1): 16-22

33. Lin WQ, Lu XY, Cao LH, Wen LL, Bai XH, Zhong ZJ. [Effects of the lung protective ventilatory strategy on proinflammatory cytokine release during one-lung ventilation]. Ai zheng = Aizheng = Chinese journal of cancer 2008; 27(8): 870-3

34. Meyhoff CS, Wetterslev J, Jorgensen LN, et al. Effect of high perioperative oxygen fraction on surgical site infection and pulmonary complications after abdominal surgery: the PROXI randomized clinical trial. Jama 2009; 302(14): 1543-50

35. Zoremba M, Dette F, Hunecke T, Braunecker S, Wulf H. The influence of perioperative oxygen concentration on postoperative lung function in moderately obese adults. European journal of anaesthesiology 2010; 27(6): 501-7

36. Hemmes SN, Serpa Neto A, Schultz MJ. Intraoperative ventilatory strategies to prevent postoperative pulmonary complications: a metaanalysis. Current opinion in anaesthesiology 2013; 26(2): 126-33

37. Güldner A K, MD T, Serpa Neto A, Canet J, Spieth P, Rocco PR, Schultz MJ, Pelosi P, Gama de Abreu M, for the PROtective VEntilation Network (PROVEnet) Intraoperative Protective Mechanical Ventilation for Prevention of Postoperative Pulmonary Complications - a Comprehensive Review of the Role of Tidal Volume, Positive End-Expiratory Pressure and Lung Recruitment Maneuvers. Anesthesiology 2015; 123(3): 692-713

38. Hemmes SN, de Abreu MG, Pelosi P, Schultz MJ. ESA Clinical Trials Network 2012: LAS VEGAS--Local Assessment of Ventilatory Management during General Anaesthesia for Surgery and its effects on Postoperative Pulmonary Complications: a prospective, observational, international, multicentre cohort study. European journal of anaesthesiology 2013; 30(5): 205-7

39. Serpa Neto A, Hemmes SN, Barbas CS, et al. Protective versus Conventional Ventilation for Surgery: A Systematic Review and Individual Patient Data Metaanalysis. Anesthesiology 2015

40. Hemmes SN, Severgnini P, Jaber S, et al. Rationale and study design of PROVHILO - a worldwide multicenter randomized controlled trial on protective ventilation during general anesthesia for open abdominal surgery. Trials 2011; 12: 111

41. Hemmes SN, Gama de Abreu M, Pelosi P, Schultz MJ. High versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial. Lancet 2014; 384(9942): 495-503

42. Hemmes SN, de Abreu MG, Pelosi P, Schultz MJ. Positive end-expiratory pressure during surgery - Authors’ reply. Lancet 2014; 384(9955): 1670-1

43. Serpa Neto A, Hemmes SN, Barbas CS, et al. Incidence of mortality and morbidity related to postoperative lung injury in patients who have undergone abdominal or thoracic surgery: a systematic review and metaanalysis. The Lancet Respiratory medicine 2014; 2(12): 1007-15

44. Dongelmans DA, Veelo DP, Paulus F, et al. Weaning automation with adaptive support ventilation: a randomized controlled trial in cardiothoracic surgery patients. Anesthesia and analgesia 2009; 108(2): 565-71

45. Dongelmans DA, Veelo DP, Binnekade JM, et al. Adaptive support ventilation with protocolized de-escalation and escalation does not accelerate tracheal extubation of patients after nonfast-track cardiothoracic surgery. Anesthesia and analgesia 2010; 111(4): 961-7

46. Dongelmans DA, Hemmes SN, Kudoga AC, Veelo DP, Binnekade JM, Schultz MJ. Positive end-expiratory pressure following coronary artery bypass grafting. Minerva anestesiologica 2012; 78(7): 790-800

47. Futier E. Positive end-expiratory pressure in surgery: good or bad? Lancet 2014; 384(9942): 472-448. Brismar B, Hedenstierna G, Lundquist H, Strandberg A, Svensson L, Tokics L. Pulmonary densities during anesthesia

with muscular relaxation--a proposal of atelectasis. Anesthesiology 1985; 62(4): 422-849. Hachenberg T, Lundquist H, Tokics L, Brismar B, Hedenstierna G. Analysis of lung density by computed tomography

before and during general anaesthesia. Acta anaesthesiologica Scandinavica 1993; 37(6): 549-55

294

50. Neumann P, Rothen HU, Berglund JE, Valtysson J, Magnusson A, Hedenstierna G. Positive end-expiratory pressure prevents atelectasis during general anaesthesia even in the presence of a high inspired oxygen concentration. Acta anaesthesiologica Scandinavica 1999; 43(3): 295-301

51. Nielsen J, Ostergaard M, Kjaergaard J, et al. Lung recruitment maneuver depresses central hemodynamics in patients following cardiac surgery. Intensive care medicine 2005; 31(9): 1189-94

52. Luecke T, Pelosi P. Clinical review: Positive end-expiratory pressure and cardiac output. Critical Care 2005; 9(6): 607-2153. Ge Y, Yuan L, Jiang X, Wang X, Xu R, Ma W. [Effect of lung protection mechanical ventilation on respiratory function in

the elderly undergoing spinal fusion]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2013; 38(1): 81-554. Severgnini P, Selmo G, Lanza C, et al. Protective mechanical ventilation during general anesthesia for open abdominal

surgery improves postoperative pulmonary function. Anesthesiology 2013; 118(6): 1307-2155. Wakabayashi K, Wilson MR, Tatham KC, O’Dea KP, Takata M. Volutrauma, but not atelectrauma, induces systemic

cytokine production by lung-marginated monocytes. Critical care medicine 2014; 42(1): e49-5756. Caironi P, Langer T, Carlesso E, Protti A, Gattinoni L. Time to generate ventilator-induced lung injury among mammals

with healthy lungs: a unifying hypothesis. Intensive care medicine 2011; 37(12): 1913-2057. Tschumperlin DJ, Oswari J, Margulies AS. Deformation-induced injury of alveolar epithelial cells. Effect of frequency,

duration, and amplitude. American journal of respiratory and critical care medicine 2000; 162(2 Pt 1): 357-6258. Garcia CS, Rocco PR, Facchinetti LD, et al. What increases type III procollagen mRNA levels in lung tissue: stress induced

by changes in force or amplitude? Respiratory physiology & neurobiology 2004; 144(1): 59-7059. Dreyfuss D, Saumon G. Role of tidal volume, FRC, and end-inspiratory volume in the development of pulmonary edema

following mechanical ventilation. The American review of respiratory disease 1993; 148(5): 1194-20360. Amato MB, Barbas CS, Medeiros DM, et al. Effect of a protective-ventilation strategy on mortality in the acute respiratory

distress syndrome. The New England journal of medicine 1998; 338(6): 347-5461. Estenssoro E, Dubin A, Laffaire E, et al. Incidence, clinical course, and outcome in 217 patients with acute respiratory

distress syndrome. Critical care medicine 2002; 30(11): 2450-662. Boissier F, Katsahian S, Razazi K, et al. Prevalence and prognosis of cor pulmonale during protective ventilation for

acute respiratory distress syndrome. Intensive care medicine 2013; 39(10): 1725-3363. Amato MB, Meade MO, Slutsky AS, et al. Driving pressure and survival in the acute respiratory distress syndrome. The

New England journal of medicine 2015; 372(8): 747-5564. Protti A, Votta E, Gattinoni L. Which is the most important strain in the pathogenesis of ventilator-induced lung injury:

dynamic or static? Current opinion in critical care 2014; 20(1): 33-865. Gattinoni L, Carlesso E, Caironi P. Stress and strain within the lung. Current opinion in critical care 2012; 18(1): 42-7

295

Nederlandse samenvatting

296

Contributing authors and affiliations

C.S. BarbasDepartment of Pneumology, Heart Institute (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BrazilDepartment of Critical Care Medicine, Hospital Israelita Albert Einstein, São Paulo, Brazil

M. BeiderlindenDepartment of Anaesthesiology, Düsseldorf University Hospital, Düsseldorf, GermanyDepartment of Anaesthesiology, Marienhospital Osnabrück, Osnabrück, Germany

M. BiehlDivision of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA

J.M. BinnekadeDepartment of Intensive Care, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands

J. CanetDepartment of Anesthesiology, Hospital Universitar I Germans Trias I Pujol, Barcelona, Spain

D.A. DongelmansDepartment of Intensive Care, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands

A. Fernandez-BustamanteDepartment of Anesthesiology, University of Colorado, Aurora, Colorado, USA

E. FutierDepartment of Anesthesiology and Critical Care Medicine, Estaing University Hospital, Clermont-Ferrand, France

O. GajicDivision of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA

M. Gama de AbreuDepartment of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany

A. Güldner Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany

G. HedenstiernaDepartment of Medical Sciences, Section of Clinical Physiology, University Hospital, Uppsala, Sweden

297

Authors and affiliations

M. HiesmayrDivision Cardiac-, Thoracic-, Vascular Anesthesia and Intensive Care, Medical University, Vienna, Austria

M.W. HollmannDepartment of Anesthesiology, Academic Medical Center, University of Amsterdam, Amsterdam, The NetherlandsLaboratory of Experimental Intensive Care and Anesthesiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands

S. Jaber Department of Critical Care Medicine and Anesthesiology (SAR B), Saint Eloi University Hospital, Montpellier, France

T. Kiss Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany

A. KozianDepartment of Anesthesiology and Intensive Care Medicine, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany

A.C. Kudoga Department of Intensive Care, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands

M. LickerDepartment of Anaesthesiology, Pharmacology and Intensive Care, Faculty of Medicine, University Hospital of Geneva, Geneva, Switzerland.

W.Q. LinState Key Laboratory of Oncology of South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China

A.D. MaslowDepartment of Anesthesiology, The Warren Alpert School of Brown University, Providence, Rhode Island

S.G. MemtsoudisDepartment of Anesthesiology, Hospital for Special Surgery, Weill Medical College of Cornell University, New York, New York, USA

P. MoineDepartment of Anesthesiology, University of Colorado, Aurora, Colorado, USA

298

T. NgDepartment of Surgery, The Warren Alpert School of Brown University, Providence, Rhode Island

D. PaparellaDepartment of Emergency and Organ Transplant (D.E.T.O.), Division of Cardiac Surgery, University of Bari Aldo Moro, Bari, Italy

P. PelosiDepartment of Surgical Sciences and Integrated Diagnostics, IRCCS San Martino IST, University of Genoa, Genoa, Italy

C. PutensenDepartment of Anesthesiology, University of Bonn, Bonn, Germany

M. RanieriAnestesiologia e Rianimazione, Dipartimento di Discipline Medico-Chirurgiche, Università di Torino, Turin, Italy

D. Reis MirandaDepartment of Intensive Care, Erasmus MC, Rotterdam, The Netherlands

P.R. RoccoLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil

F. ScavonettoDepartment of Anesthesiology and Anesthesia Clinical Research Unit, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA

T. SchillingDepartment of Anesthesiology and Intensive Care Medicine, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany

W. SchmidDepartment of Cardiothoracic and Vascular Anesthesia and Intensive Care, Medical University, Vienna, Austria

M.J. SchultzDepartment of Intensive Care, Academic Medical Center, University of Amsterdam, Amsterdam, The NetherlandsLaboratory of Experimental Intensive Care and Anesthesiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands

G. SelmoDepartment of Environment, Health and Safety, University of Insubria, Varese, Italy

299

Authors and affiliations

A. Serpa NetoDepartment of Intensive Care, Academic Medical Center, University of Amsterdam, Amsterdam, The NetherlandsDepartment of Pneumology, Heart Institute (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BrazilDepartment of Critical Care Medicine, Hospital Israelita Albert Einstein, São Paulo, Brazil

P. Severgnini Department of Environment, Health and Safety, University of Insubria, Varese, Italy

P. SpiethDepartment of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany

J. Sprung Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA

S. SundarDepartment of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA

D. TalmorDepartment of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA

T. TreschanDepartment of Anaesthesiology, Düsseldorf University Hospital, Düsseldorf, Germany

E.M. TschernkoDivision Cardiac-, Thoracic-, Vascular Anesthesia and Intensive Care, Medical University, Vienna, Austria

C. UnzuetaDepartment of Anaesthesiology, Hospital de Sant Pau, Barcelona, Spain

D.P. Veelo Department of Anesthesiology, Academic Medical Center, University of Amsterdam, Amsterdam, The NetherlandsLaboratory of Experimental Intensive Care and Anesthesiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands

T.N. WeingartenDepartment of Anesthesiology and Anesthesia Clinical Research Unit, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA

300

E.K. WolthuisDepartment of Intensive Care, Academic Medical Center, University of Amsterdam, Amsterdam, The NetherlandsLaboratory of Experimental Intensive Care and Anesthesiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands

H. WriggeDepartment Anesthesiology and Intensive Care Medicine, University of Leipzig, Leipzig, Germany

301

Authors and affiliations

302

Publications SNT Hemmes

Güldner A, Kiss T, Serpa Neto A, Hemmes SN, Canet J, Spieth PM, Rocco PR, Schultz MJ, Pelosi P, de Abreu MG.Intraoperative Protective Mechanical Ventilation for Prevention of Postoperative Pulmonary Complications: A Comprehensive Review of the Role of Tidal Volume, Positive End-expiratory Pressure, and Lung Recruitment Maneuvers.Anesthesiology 2015; 122(3): 631-46

Serpa Neto A, Hemmes SN, Barbas CS, Beiderlinden M, Biehl M, Binnekade JM, Canet J, Fernandez-Bustamante A, Futier E, Gajic O, Hedenstierna G, Hollmann MW, Jaber S, Kozian A, Licker M, Lin WQ, Maslow AD, Memtsoudis SG, Reis Miranda D, Moine P, Ng T, Paparella D, Putensen C, Ranieri M, Scavonetto F, Schilling T, Schmid W, Selmo G, Severgnini P, Sprung J, Sundar S, Talmor D, Treschan T, Unzueta C, Weingarten TN, Wolthuis EK, Wrigge H, Gama de Abreu M, Pelosi P, Schultz MJ.Protective versus Conventional Ventilation for Surgery: A Systematic Review and Individual Patient Data Metaanalysis.Anesthesiology 2015; May 15 [ePub ahead of print]

Serpa Neto A, Hemmes SN, Barbas CS, Beiderlinden M, Fernandez-Bustamante A, Futier E, Hollmann MW, Jaber S, Kozian A, Licker M, Lin WQ, Moine P, Scavonetto F, Schilling T, Selmo G, Severgnini P, Sprung J, Treschan T, Unzueta C, Weingarten TN, Wolthuis EK, Wrigge H, Gama de Abreu M, Pelosi P, Schultz MJ; PROVE Network investigators.Incidence of mortality and morbidity related to postoperative lung injury in patients who have undergone abdominal or thoracic surgery: a systematic review and metaanalysis.Lancet Respir Med 2014;2(12):1007-15

Hemmes SN, de Abreu MG, Pelosi P, Schultz MJ.Positive end-expiratory pressure during surgery - Authors’ reply.Lancet 2014;8;384(9955):1670-1

PROVE Network Investigators for the Clinical Trial Network of the European Society of Anaesthesiology, Hemmes SN, Gama de Abreu M, Pelosi P, Schultz MJ.High versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial.Lancet 2014;9;384(9942):495-503

Bakker OG, Hemmes SN, Backes Y, Determann RM, Schultz MJ.SuPAR in pleural fluid may function as a biological marker for infection in critically ill patients with pleural effusions.J Infect 2014;68(6):607-9

Serpa Neto A, Hemmes SN, de Abreu MG, Pelosi P, Schultz MJ; PROVE Network investigators.Protocol for a systematic review and individual patient data metaanalysis of benefit of so-called lung-protective ventilation settings in patients under general anesthesia for surgery.Syst Rev 2014;2;3:2

303

Publications

Hegeman MA, Hemmes SN, Kuipers MT, Bos LD, Jongsma G, Roelofs JJ, van der Sluijs KF, Juffermans NP, Vroom MB, Schultz MJ.The extent of ventilator-induced lung injury in mice partly depends on duration of mechanical ventilation.Crit Care Res Pract;2013:435236

Hemmes SN, Paulus F, Schultz MJ.From the dark side of ventilation toward a brighter look at lungs.Crit Care Med 2013;41(5):1376-7

Hemmes SN, de Abreu MG, Pelosi P, Schultz MJ.ESA Clinical Trials Network 2012: LAS VEGAS--Local Assessment of Ventilatory Management during General Anaesthesia for Surgery and its effects on Postoperative Pulmonary Complications: a prospective, observational, international, multicentre cohort study.Eur J Anaesthesiol 2013;30(5):205-7

Hemmes SN, Serpa Neto A, Schultz MJ.Intraoperative ventilatory strategies to prevent postoperative pulmonary complications: a metaanalysis.Curr Opin Anaesthesiol;2013;26(2):126-33.

Dongelmans DA, Hemmes SN, Kudoga AC, Veelo DP, Binnekade JM, Schultz MJ.Positive end-expiratory pressure following coronary artery bypass grafting.Minerva Anestesiol 2012;78(7):790-800

Hemmes SN, Severgnini P, Jaber S, Canet J, Wrigge H, Hiesmayr M, Tschernko EM, Hollmann MW, Binnekade JM, Hedenstierna G, Putensen C, de Abreu MG, Pelosi P, Schultz MJ.Rationale and study design of PROVHILO - a worldwide multicenter randomized controlled trial on protective ventilation during general anesthesia for open abdominal surgery.Trials 2011;6;12:111

Submitted manuscriptsHemmes SN, Gama de Abreu M, Pelosi P, Schultz MJ for the The LAS VEGAS Investigators for PROVE Network*, and the Clinical Trial Network of the European Society of AnaesthesiologyIntraoperative Ventilation Strategies and Patient Outcomes Following Surgery: an International Observational Study (LAS VEGAS).*PROVE Network: the PROtective VEntilation Network

304

PhD Portfolio

PhD student: S.N.T. HemmesSupervisors: Prof. dr. M.J. Schultz and prof. dr. dr. M.W. HollmannPhD period: September 2010 – December 2015

PhD Training Year ECTs

Courses

AMC World of Science 2010 0.7

Crash Course Biochemistry 2010 0.4

Basiscursus regelgeving klinisch onderzoek (BROK) 2011 0.9

Laboratory animals 2011 3.9

Evidence Based Searching 2011 0.1

Clinical Epidemiology 2011 0.6

Systematic Reviews 2011 0.3

Clinical Data Management 2011 0.2

Oral Presentation 2012 0.8

Practical Biostatistics 2012 1.1

Entrepreneurship in Health and Life Sciences 2012 0.4

Presentations

Study design of the PROVHILO trial - Protective Ventilation using High versus Low PEEP. Oral presentation and meeting at Euroanesthesia 2011 0.3

Do Soluble Mediators Released In The Airways During Mechanical Ventilation Cause Ventilator-Induced Lung Injury? Poster at the American Thoracic Society International Conference

2012 0.5

Effects of manual hyperinflation on lung aeration in direct and indirect ARDS. Poster at the Annual Congress of the European Society of Intensive Care Medicine

2013 0.5

Study design and first results of (LAS VEGAS) Local ASsessment of VEntilatory Management During General Anesthesia for Surgery and effects on Postoperative Pulmonary Complications: a Prospective Observational International Multi–center Cohort Study. Oral presentation and meeting at Euroanesthesia

2014 0.3

305

PhD Portfolio

International conferences

Euroanesthesia 2011 0.75

American Thoracic Society International Conference 2012 1.0

Euroanesthesia 2012 0.75

Annual Congress of the European Society of Intensive Care Medicine 2012 0.75

Euroanesthesia 2013 0.75

Annual Congress of the European Society of Intensive Care Medicine 2013 0.75

Euroanesthesia 2014 0.75

Euroanesthesia 2015 0.75

Other activities

Intensive Care Journal Club (monthly) 2010 - 2013 3

Intensive Care research meeting (weekly) 2010 – 2013 13.5

Laboratory of Experimental Intensive Care and Anesthesiology (LEICA) research meeting (weekly) 2010 – 2013 13.5

Anesthesiology evening seminars (monthly) 2010 - 2015 4

PhD Training Year ECTs

Teaching

Student coaching and mentoring

Research Internship, Medicine, University of Amsterdam 2012 1.0

Research Internship, Medicine, University of Amsterdam 2012 1.0

Bachelor thesis, Medicine, University of Amsterdam 2012 1.0

Research Internship, Medicine, University of Amsterdam 2013 1.0

Lecturing

306

Can intra-operative mechanical ventilation harm our patients? Oral presentation at the Laboratory of Experimental Intensive Care and Anesthesiology (LEICA) symposium

2013 0.5

Met welk Tidal Volume moeten we ventileren? Yearly training course of Anesthesiologists and Intensivists from The Netheralnds and Belgium. CEEA Course, Antwerpen

Mar&Okt2015 2

High levels of PEEP – Tipping the balance? Oral presentation at AMC symposium, The Update on Intraoperative Ventilation Dec 2015 0.5

Parameters of esteem

Grants

European Society of Anesthesiology Research Grant for PROVHILO trial 2011

European Society of Anesthesiology Clinical Trial Network Grant for LAS VEGAS trial 2012

Awards

ATS International Trainee Travel Award 2012

Ritsema van Eck award, Nederlandse Vereniging van Anesthesiologie, 1st place 2015

DRÄGER Prize in Anaesthesia and Intensive Care Medicine 2015

AMC PhD Publication 2014 Award, 2nd place 2015

307

PhD Portfolio

308

Curriculum Vitae

Sabrine Hemmes was born on 23 March 1982 in Hilversum, where she spent the first 5 years of her life. After a 3-year period in Atlanta, U.S.A. with her parents, sister and brother, the family came back to Blaricum in 1990. Ten years later, Sabrine graduated from high-school at the Gemeentelijk Gymnasium in Hilversum. Afterwards she spent a year abroad, learning the Spanish language and offering volunteer work to burn-patients in Peru. In this gap-year, Sabrine decided to study Medicine. During her travels through South-America she participated in the ‘decentrale selectie’ of the AMC in Amsterdam with success. In 2011 she commenced with her Medical studies at the Academic Medical Center, University of Amsterdam. During her studies she had a special interest in Tropical Medicine. She completed a summer school in infectious diseases in Egypt and performed her first scientific research on malaria in Malawi. After obtaining her doctors degree in 2008 she left for Uganda to work as a doctor in rural settings for several months. She gained further medical experience at the Surgical Department of the Slotervaartziekenhuis in Amsterdam and the Intensive Care of the Rijnland Ziekenhuis in Leiderdorp. In 2010 she applied to and was accepted for the specialization in Anaesthesiology at the AMC. Meanwhile, Sabrine had become acquainted with prof. dr. Marcus J. Schultz (intensivist), who supervised the work presented in this thesis together with prof. dr. dr. Markus W. Hollmann (anaesthesiologist). In 2013 Sabrine started her traineeship as anaesthesiologist, which she will complete in 2018. She will perform part of her residency in Intensive Care in 2016 in Melbourne, Australia. Next to her clinical activities, Sabrine is still actively involved in several running research projects related to her PhD projects.

309

Curriculum Vitae

310

Acknowledgements

On the road to my doctorate, countless people have helped me and played an essential role. I would like to thank them all, and some in particular.

First of all, the patients who selflessly participated in the several trials and studies. Secondly, all collaborating investigators for their hard work and efforts.Thirdly, all anaesthetic, surgical, out-patient clinic and ward staff, who gave us the opportunity to perform our clinical studies.

I am thankful to my promotores prof. Marcus Schultz and prof. Markus Hollmann:Marcus, for believing in me scientifically, by giving me the opportunity to start on this PhD project. Initiating an international multicentre trial was a new bold adventure for the both of us, which turned out to be a great success and a starting-point for many other international projects.Markus, for believing in me clinically, by giving me time off from clinical duties to focus on my research. Furthermore thank you for your scientific and emotional support; I always had the feeling that you ‘had my back’.

I would like to thank the other members of my doctorate committee for reviewing my thesis and attending my doctoral defence: prof. dr. L.P.H.J. Aarts, prof. dr. C. Boer, prof. dr. M.A. Boermeester, prof. dr. G. Cinnella, prof. dr. M.M. Levi, prof. dr. W.S. Schlack en prof. dr. M.B. Vroom.

Many thanks and warmest wishes to the two professors and one professor-to-be with whom I have collaborated so closely together with Marcus, and with whom I have shared literally thousands of emails: prof. Paolo Pelosi, prof. Marcelo Gama de Abreu and dr. Ary Serpa Neto. The amount of passion and work that you all put into your many simultaneously running studies and into PROVEnet is mindboggling and inspiring.

The European Society of Anaesthesiology for generously supporting several of our studies and for believing in our ideas. In specific Brigitte, Sandrine and Benoit for their endless efforts and support in the execution of PROVHILO and LAS VEGAS. It has been a tremendous job and a great achievement! We could (really!) not have done it without you. Above all it was a great pleasure to work with you.

Mijn kamergenoten gedurende mijn eerste 3 promotiejaren: Luuk en Lieuwe. Jullie waren mijn wetenschappelijk brein, klankbord en geweten. Zonder jullie had ik dit dankwoord nu niet geschreven. Luuk, veel dank voor jouw eindeloos geduld, luisterend oor en diplomatieke adviezen. Niet alleen op wetenschappelijk vlak, maar ook op het gebied van huizenkoperij, belasting en persoonlijk vlak.Lieuwe, ik had graag gewild dat jij mijn co-promotor had kunnen zijn. Ik heb het geprobeerd! Je hebt het zeker verdiend; ik heb zoveel met jou gebrainstormd over mijn onderzoeken en over beademing in het algemeen. Het stelde mij altijd gerust, dat -mocht ik onder een tram komen- jij feilloos al mijn onderzoek zou kunnen overnemen (gelukkig had jij ook al mijn wachtwoorden). Jullie waren mijn belangrijkste steunpilaren binnen mijn promotie. Balen dat jullie beiden niet aan mijn zijde kunnen staan op ‘le moment suprême’.

311

Acknowledgements

Aline, jij hoort hier eigenlijk ook bij. We hebben veel te kort van je vrolijkheid en wijsheid mogen genieten; jij maakte onze drie-eenheid tot een evenwichtig kwartet.

Thanks to all my other co-PhD’s from the Intensive Care and LEICA: Roos, Marleen, Charlotte, Friso, Hamid, Ilse, Daniel, Djai, Gezina, Robert, Lonneke, Maryse, Esther, Laura, Gerie, Rianne, Frank, Hendrik en Hemmik for the great times we have had. I always considered the ICU/LEICA research-group as the greatest PhD-group of the AMC.Furthermore, I would like to thank the other researchers of the ICU: Frederique, Jan, Marjon, Annelou, Tineke, Mark, Nicole, Janneke, Dave, Marcella, Thomas, Wim, Mary-Anne, Erica, and from the LEICA: Anita, Gearstje, Jessica en Koen. Specifically Jan for all the hours of work on the impossible data-exports, Annelou for performing the PROVHILO monitoring abroad, Anita for all her help and gezelligheid in the lab, and Frederique for the great collaboration and the long talks about PhD-life and life outside research.

Many thanks to all my great colleagues, supervisors and nurse anaesthetists from the Anaesthesia Departments of both the AMC and OLVG, and to dr. Peter de Haan in specific. I have learnt so much from all of you the last two years. It has been a great pleasure to work by your side!

Love and thanks to all my fantastic and inspiring friends, who give so much joy and richness to my life. The last two years I’ve seen far to little of you guys… Change is coming!

My dear paranimfs Jet and Pieter Roel; it was an honour for me to be paranimf at both your defences. Now I am very proud to have such brilliant people by my side during my thesis defence. Jet, you are one of the warmest, most supportive people I know. And lucky me; I had you close during my PhD course, as we did our doctoral programmes at the same time in the same hospital! I hope to enjoy your friendship the rest of my life. Pieter Roel, we started off on our PhD in room G3-227, where we shared the same humour and our love for music. You soon left to complete your ICU fellowship, but it has always surprised me that we developed such a great friendship in such a short time. Lots of love to my family, who provided a stable, warm home and for the greatest part made me into the person I am today. They gave me endless support during my PhD project, even though they maybe did not always completely comprehend the mysteries of Medical Science. “Okay, so if I understand correctly... On a scale from Donald Duck to Times Magazine, you just published in the Times!”

Lieve Hans, jij maakt mij tot een beter mens.


Recommended